EP3664820B1 - Methods for producing genetically engineered cell compositions and related compositions - Google Patents

Methods for producing genetically engineered cell compositions and related compositions Download PDF

Info

Publication number
EP3664820B1
EP3664820B1 EP18760159.6A EP18760159A EP3664820B1 EP 3664820 B1 EP3664820 B1 EP 3664820B1 EP 18760159 A EP18760159 A EP 18760159A EP 3664820 B1 EP3664820 B1 EP 3664820B1
Authority
EP
European Patent Office
Prior art keywords
cells
instances
naïve
antigen
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP18760159.6A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP3664820A1 (en
Inventor
Mirna MUJACIC
Ayu RAHARDJO
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Juno Therapeutics Inc
Original Assignee
Juno Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Juno Therapeutics Inc filed Critical Juno Therapeutics Inc
Publication of EP3664820A1 publication Critical patent/EP3664820A1/en
Application granted granted Critical
Publication of EP3664820B1 publication Critical patent/EP3664820B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464416Receptors for cytokines
    • A61K39/464417Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR], CD30
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • G01N33/56972White blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/11Coculture with; Conditioned medium produced by blood or immune system cells
    • C12N2502/1114T cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13041Use of virus, viral particle or viral elements as a vector
    • C12N2740/13043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70514CD4
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70517CD8

Definitions

  • the present disclosure relates to methods and compositions for generating engineered cells, such as cells expressing a recombinant receptor, including methods involving stimulation and/or engineering of an input composition having a defined ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells.
  • the methods can be used to engineer T cells with genetically engineered receptors, such as genetically engineered antigen receptors such as engineered (recombinant) TCRs and chimeric antigen receptors (CARs), or other recombinant chimeric receptors.
  • genetically engineered receptors such as genetically engineered antigen receptors such as engineered (recombinant) TCRs and chimeric antigen receptors (CARs), or other recombinant chimeric receptors.
  • CARs chimeric antigen receptors
  • Various methods are available for preparing cells for therapeutic use and administering the cells.
  • methods are available for preparing cells, including T cells, for engineering and cell therapy, including methods involving depletion of or enrichment for certain sub-populations.
  • Improved methods are needed, for example, to reduce toxicity associated with certain adoptive cell therapy administrations, to improve the manufacturing process, to allow improved administration, and/or to reduce cost or other resources.
  • Provided are methods, cells, compositions, kits, and systems that meet such needs.
  • WO2012/129514A1 describes methods and compositions for immunotherapy.
  • a method for generating a cell composition including (a) combining a first cell composition containing na ⁇ ve-like CD4+ T cells with a second cell composition containing na ⁇ ve-like CD8+ T cells to produce an input cell composition in which the ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells is between or about between 0.8:1 and 2.2:1, inclusive,
  • the method includes determining the number, number per volume, number per weight, and/or percentage of the na ⁇ ve-like CD4+ T cells in the first cell composition and/or the number, number per volume, number per weight, and/or percentage of the na ⁇ ve-like CD8+ T cells in the second composition. In some cases, prior to the combining, the method includes determining the number, number per volume, number per weight, and/or percentage of the na ⁇ ve-like CD4+ T cells and/or the number , number per volume, number per weight, and/or percentage of na ⁇ ve-like CD8+ T cells in the biological sample from the subject.
  • the ratio of the na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells in the input composition is adjusted or altered compared to the ratio of the na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells in the biological sample from the subject.
  • the method includes determining the number, number per volume, number per weight, and/or percentage of na ⁇ ve-like CD4+ T cells and na ⁇ ve-like CD8+ T cells in a biological sample obtained from a subject or in one or more samples derived therefrom; and producing an input composition containing CD4+ T cells and CD8+ T cells in which the ratio of the na ⁇ ve-like CD4+ T cells to naive-like CD8+ T cells is between or about between 2.2:1 to 0.8:1, inclusive, wherein said ratio in the input composition is adjusted or altered compared to the ratio of the na ⁇ ve-like CD4+ T cells to naive-like CD8+ T cells in the biological sample from the subject.
  • the method includes contacting the input composition with an agent containing a nucleic acid molecule encoding a recombinant receptor under conditions to introduce the nucleic acid encoding the recombinant receptor into cells in the input composition.
  • the method includes contacting an input composition containing na ⁇ ve-like CD4+ T cells and na ⁇ ve-like CD8+ T cells from a biological sample from a subject with an agent containing a nucleic acid molecule encoding a recombinant receptor under conditions to introduce the nucleic acid encoding the recombinant receptor into cells in the composition, wherein the ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells present in the input composition is between or about between 0.8:1 and 2.2:1, inclusive.
  • the method further includes stimulating the cells, prior to, during and/or subsequent to said contacting, wherein stimulating includes incubating the cells in the presence of one or more stimulating agents, wherein stimulating results in activation and/or proliferation of the cells.
  • the method includes combining a first cell composition containing na ⁇ ve-like CD4+ T cells with a second cell composition containing na ⁇ ve-like CD8+ T cells to produce an input cell composition in which the ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells is between or about between 0.8:1 and 2.2:1, inclusive; contacting the input composition with an agent containing a nucleic acid molecule encoding a recombinant receptor under conditions to introduce the nucleic acid encoding the recombinant receptor into cells in the composition; and stimulating the cells, prior to, during and/or subsequent to said contacting, wherein stimulating includes incubating the cells in the presence of one or more stimulating agents, wherein stimulating results in activation and/or proliferation of the cells.
  • the na ⁇ ve-like CD4+ cells are surface positive for CD45RA and CCR7; (ii) are surface positive CD27 and CCR7; or (iii) are surface positive for CCR7 and surface negative for CD62L; and the na ⁇ ve-like CD8+ cells (i) are surface positive for CD45RA and CCR7; (ii) are surface positive CD27 and CCR7; or (iii) are surface positive for CCR7 and surface negative for CD62L.
  • the na ⁇ ve-like CD4+ and/or the na ⁇ ve-like CD8+ cells are surface positive for a marker selected from the group consisting of CD45RA, CD27, CD28, CD62L, and CCR7; and/or are surface negative for a marker selected from the group consisting of CD25, CD45RO, CD56, KLRG1; and/or have low expression of CD95; and/or are negative for intracellular expression of a cytokine selected from the group consisting of IL-2, IFN- ⁇ , IL-4, IL-10.
  • the na ⁇ ve-like CD4+ and/or the na ⁇ ve-like CD8+ cells are surface positive for a T cell activation marker selected from the group consisting of CD45RA, CD27, CD28, and CCR7; and/or are surface negative for a marker selected from the group consisting of CD45RO, CD56, KLRG1; and/or have low expression of CD95.
  • the na ⁇ ve-like CD4+ and/or the na ⁇ ve-like CD8+ cells are surface positive for CD45RA and CCR7.
  • the na ⁇ ve-like CD4+ and the na ⁇ ve-like CD8+ cells are surface positive for CD45RA, CD27 and CCR7 and are surface negative for CD45RO.
  • the number, number per volume, number per weight, and/or percentage of na ⁇ ve-like CD4+ T cells and/or the number, number per volume, number per weight, and/or percentage of na ⁇ ve-like CD8+ T cells is determined by flow cytometry.
  • the ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells has been adjusted compared to the ratio of the na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells in a biological sample from the subject.
  • the biological sample is or is obtained from a blood, plasma or serum sample.
  • the biological sample is or includes a whole blood sample, a buffy coat sample, a peripheral blood mononuclear cells (PBMC) sample, an unfractionated T cell sample, a lymphocyte sample, a white blood cell sample, an apheresis product, or a leukapheresis product.
  • the biological sample is or is obtained from an apheresis or leukapheresis sample.
  • the subject is a human subject.
  • the input composition contains a ratio of na ⁇ ve-like CD4+ cells to na ⁇ ve-like CD8+ cells of between or about between 0.8:1 and 2.0:1, 0.8:1 and 1.6:1, 0.8:1 and 1.4:1, 0.8:1 and 1.2:1 or 1.0:1 and 1.2:1, each inclusive.
  • the input composition includes a ratio of na ⁇ ve-like CD4+ cells to na ⁇ ve-like CD8+ cells of or about 1.5:1, 1.4:1, 1.3:1, 1.2:1, 1.1:1, or 1.0:1.
  • the input composition contains a ratio of na ⁇ ve-like CD4+ cells to na ⁇ ve-like CD8+ cells of or about 1.1:1.
  • the input composition contains from or from about 1 ⁇ 10 7 to 5 ⁇ 10 9 total cells or total T cells, from or from about 5 ⁇ 10 7 to 1 ⁇ 10 9 total cells or total T cells, from or from about 1 ⁇ 10 8 to 5 ⁇ 10 8 total cells or total T cells, or from or from about 2 ⁇ 10 8 to 5 ⁇ 10 8 total cells or total T cells, or of viable populations of any of the foregoing.
  • the input composition contains at least or at least about 1 ⁇ 10 8 , 2 ⁇ 10 8 , 3 ⁇ 10 8 , 4 ⁇ 10 8 , or 5 ⁇ 10 8 total cells or total T cells or a viable population of any of the foregoing.
  • the one or more stimulating agent is capable of activating T cells, CD4+ T cells and/or CD8+ T cells; is capable of inducing a signal through a TCR complex; and/or is capable of inducing proliferation of T cells, CD4+ T cells and/or CD8+ T cells.
  • the one or more stimulating agent contains a primary agent that binds to a member of a TCR complex, optionally that specifically binds to CD3.
  • the one or more stimulating agent further contains a secondary agent that specifically binds to a T cell costimulatory molecule.
  • the costimulatory molecule is selected from the group consisting of CD28, CD137 (4-1-BB), OX40, or ICOS.
  • the primary and secondary agents contain antibodies, optionally wherein the one or more stimulating agent includes incubation with an anti-CD3 antibody and an anti-CD28 antibody.
  • the one or more stimulating agents are present on the surface of a solid support, optionally a bead.
  • the one or more stimulating agent are present on the surface of a bead and the bead is a paramagnetic bead.
  • the one or more one stimulating agent is selected from the group consisting of CD3-binding molecules; CD28-binding molecules; recombinant IL-2; recombinant IL-15; and recombinant IL-7, a vaccine containing an antigen specifically recognized by the antigen receptor, and an anti-idiotype antibody that specifically binds the antigen receptor or combinations thereof.
  • the incubation is carried out for 2 to 15 days, 2 to 12 days, 2 to 12 days, 2 to 8 days, 2 to 6 days, 2 to 4 days, 4 to 12 days, 4 to 10 days, 4 to 8 days, 4 to 6 days, 6 to 12 days, 6 to 10 days, 6 to 8 days, 8 to 12 days, 8 to 10 days, or 10 to 12 days. In some cases, the incubation is carried out for at least or about at least or 4 days, 6 days, 8 days, 10 days or 12 days.
  • the agent containing the nucleic acid molecule is a viral vector or is a transposon. In some cases, the agent containing the nucleic acid molecule is a viral vector and the viral vector is a retroviral vector. In some examples, the viral vector is a lentiviral vector or a gammaretroviral vector.
  • the recombinant receptor is capable of binding to a target antigen that is associated with, specific to, and/or expressed on a cell or tissue of a disease, disorder or condition.
  • the disease, disorder or condition is an infectious disease or disorder, an autoimmune disease, an inflammatory disease, or a tumor or a cancer.
  • the target antigen is a tumor antigen.
  • the target antigen is selected from among ROR1, B cell maturation antigen (BCMA), carbonic anhydrase 9 (CAIX), tEGFR, Her2/neu (receptor tyrosine kinase erbB2), CD19, CD20, CD22, mesothelin, CEA, and hepatitis B surface antigen, anti-folate receptor, CD23, CD24, CD30, CD33, CD38, CD44, EGFR, epithelial glycoprotein 2 (EPG-2), epithelial glycoprotein 40 (EPG-40), EPHa2, erb-B2, erb-B3, erb-B4, erbB dimers, EGFR vIII, folate binding protein (FBP), FCRL5, FCRH5, fetal acetylcholine receptor, GD2, GD3, G Protein Coupled Receptor 5D (GPCR5D, HMW-MAA, IL-22R-alpha, kinase insert domain receptor
  • the target antigen is selected from among Receptor Tyrosine Kinase Like Orphan Receptor 1 (ROR1), B cell maturation antigen (BCMA), carbonic anhydrase 9 (CA9, also known as CAIX or G250), Her2/neu (receptor tyrosine kinase erbB2), CD19, CD20, CD22, mesothelin (MSLN), carcinoembryonic antigen (CEA), and hepatitis B surface antigen, anti-folate receptor, CD23, CD24, CD30, CD33, CD38, CD44, chondroitin sulfate proteoglycan 4 (CSPG4), EGFR, epithelial glycoprotein 2 (EPG-2), epithelial glycoprotein 40 (EPG-40), ephrin receptor A2 (EPHa2), Her3 (erb-B3), Her4 (erb-B4), erbB dimers, type III epidermal growth factor receptor mutation (ROR1)
  • Antigens targeted by the receptors include antigens associated with a B cell malignancy, such as any of a number of known B cell marker.
  • the antigen is or includes CD20, CD19, CD22, ROR1, CD45, CD21, CD5, CD33, Igkappa, Iglambda, CD79a, CD79b or CD30.
  • the antigen is or includes a pathogen-specific or pathogen-expressed antigen.
  • the antigen is a viral antigen (such as a viral antigen from HIV, HCV, HBV, etc.), bacterial antigens, and/or parasitic antigens.
  • the recombinant receptor is or contains a functional non-TCR antigen receptor or a TCR or antigen-binding fragment thereof.
  • the recombinant receptor is a chimeric antigen receptor (CAR).
  • the chimeric antigen receptor contains an extracellular domain containing an antigen-binding domain.
  • the antigen-binding domain is or contains an antibody or an antibody fragment thereof, which optionally is a single chain fragment.
  • the fragment contains antibody variable regions joined by a flexible linker.
  • the fragment contains an scFv.
  • the chimeric antigen receptor further contains a spacer and/or a hinge region. In some embodiments, the chimeric antigen receptor contains an intracellular signaling region. In some cases, the intracellular signaling region contains an intracellular signaling domain. In some embodiments, the intracellular signaling domain is or contains a primary signaling domain, a signaling domain that is capable of inducing a primary activation signal in a T cell, a signaling domain of a T cell receptor (TCR) component, and/or a signaling domain contianing an immunoreceptor tyrosine-based activation motif (ITAM). In some embodiments, the intracellular signaling domain is or contains an intracellular signaling domain of a CD3 chain, optionally a CD3-zeta (CD3 ⁇ ) chain, or a signaling portion thereof.
  • TCR T cell receptor
  • ITAM immunoreceptor tyrosine-based activation motif
  • the CAR comprises an scFv specific for the antigen, a transmembrane domain, a cytoplasmic signaling domain derived from a costimulatory molecule, which optionally is or comprises a 4-1BB, and a cytoplasmic signaling domain derived from a primary signaling ITAM-containing molecule, which optionally is or comprises a CD3zeta signaling domain and optionally further comprises a spacer between the transmembrane domain and the scFv;
  • the CAR comprises, in order, an scFv specific for the antigen, a transmembrane domain, a cytoplasmic signaling domain derived from a costimulatory molecule, which optionally is or comprises a 4-1BB signaling domain, and a cytoplasmic signaling domain derived from a primary signaling ITAM-containing molecule, which optionally is a CD3zeta signaling domain; or the CAR comprises, in order, an scFv specific for the antigen, a
  • the chimeric antigen receptor further contains a transmembrane domain disposed between the extracellular domain and the intracellular signaling region.
  • the intracellular signaling region further contains a costimulatory signaling region.
  • the costimulatory signaling region contains an intracellular signaling domain of a T cell costimulatory molecule or a signaling portion thereof.
  • the costimulatory signaling region contains an intracellular signaling domain of a CD28, a 4-1BB or an ICOS or a signaling portion thereof.
  • the costimulatory signaling region is between the transmembrane domain and the intracellular signaling region.
  • the subject has a disease or condition, optionally wherein the recombinant receptor specifically recognizes or specifically bind to an antigen associated with, or expressed or present on cells of, the disease or condition.
  • the method produces an output composition in which the ratio of recombinant receptor-expressing CD4+ T cells to recombinant receptor-expressing CD8+ T cells, optionally the ratio of viable cells thereof, varies by no more than 20% or no more than 10% or no more than 5% from an average of said ratio in a plurality of T cell compositions produced by the method and/or varies from such average by no more than one standard deviation.
  • the method produces an output composition in which the ratio of recombinant receptor-expressing CD4+ T cells to recombinant receptor-expressing CD8+ T cells, optionally the ratio of viable cells thereof, is between at or about 0.5:1 and 2:1 or 0.8:1 and 1.6:1 or 1:1 and 1.5:1, each inclusive.
  • the ratio of recombinant receptor-expressing CD4+ T cells to recombinant receptor-expressing CD8+ T cells, optionally the ratio of viable cells thereof, in the output composition is or is about 1.2:1, 1.1:1, 1:1, 0.9:1, or 0.8:1. In some cases, the ratio of recombinant receptor-expressing CD4+ T cells to recombinant receptor-expressing CD8+ T cells, optionally the ratio of viable cells thereof, in the output composition is or is about 1:1.
  • the viable cells contain cells that are apoptotic marker negative (-), optionally wherein the apoptotic marker is Annexin V or active Caspase 3.
  • the method is performed in vitro or ex vivo.
  • an output composition produced by the method of the invention. Also provided is a pharmaceutical composition containing the output composition described. In some embodiments, the pharmaceutical composition further contains a pharmaceutical carrier.
  • Disclosed herein is a method of treatment including administering to a mammalian subject an output composition produced by any of the described methods or any of the described pharmaceutical compositions.
  • the cells may be derived from the subject to which the cells are administered.
  • the na ⁇ ve-like CD4+ and/or the na ⁇ ve-like CD8+ cells are surface positive for CD45RA and CCR7. In some embodiments of the provided methods, the na ⁇ ve-like CD4+ and/or the na ⁇ ve-like CD8+ cells are surface positive for CD27 and CCR7. In particular embodiments of the provided methods, the na ⁇ ve-like CD4+ and the na ⁇ ve-like CD8+ cells are surface positive for CD45RA, CD27 and CCR7 and are surface negative for CD45RO.
  • the na ⁇ ve-like CD4+ and/or the na ⁇ ve-like CD8+ cells are surface positive for CCR7 and surface negative for CD62L.
  • the input composition comprises a ratio of na ⁇ ve-like CD4+ cells to na ⁇ ve-like CD8+ cells that are surface positive for CD45RA and CCR7 of or about 1.1:1.
  • the input composition comprises a ratio of na ⁇ ve-like CD4+ cells to na ⁇ ve-like CD8+ cells that are surface positive for CD45RA and CD27 of or about 1.69:1.
  • the input cell composition comprises a ratio of na ⁇ ve-like CD4+ cells to na ⁇ ve-like CD8+ cells that are surface positive for CD27 and CCR7 of or about 1.69:1.
  • the method comprises: combining a first cell composition comprising CCR7+CD45RA+CD4+ T cells with a second cell composition comprising CCR7+CD45RA+CD8+ T cells to produce an input cell composition in which the ratio of CCR7+CD45RA+CD4+ T cells to CCR7+CD45RA+CD8+ T cells is between or about between 0.8:1 and 2.2:1, inclusive.
  • the first cell composition is produced by isolating CD4+ T cells from a biological sample obtained from a subject and/or the second cell composition is produced by isolating CD8+ T cells from the biological sample obtained from the subject.
  • the method comprises determining the number, number per volume, number per weight, and/or percentage of the CCR7+CD45RA+CD4+ T cells in the first cell composition and/or the number, number per volume, number per weight, and/or percentage of the CCR7+CD45RA+CD8+ T cells in the second composition.
  • the method comprises determining the number, number per volume, number per weight, and/or percentage of the CCR7+CD45RA+CD4+ T cells and/or the number, number per volume, number per weight, and/or percentage of CCR7+CD45RA+CD8+ T cells in the biological sample from the subject.
  • the ratio of the CCR7+CD45RA+CD4+ T cells to CCR7+CD45RA+CD8+ T cells in the input composition is adjusted or altered compared to the ratio of the CCR7+CD45RA+CD4+ T cells to CCR7+CD45RA+CD8+ T cells in the biological sample from the subject.
  • the method comprises: determining the number, number per volume, number per weight, and/or percentage of CCR7+CD45RA+CD4+ T cells and CCR7+CD45RA+CD8+ T cells in a biological sample obtained from a subject or in one or more samples derived therefrom; and producing an input composition comprising CD4+ T cells and CD8+ T cells in which the ratio of the CCR7+CD45RA+CD4+ T cells to naive-like CD8+ T cells is between or about between 2.2:1 to 0.8:1, inclusive, wherein said ratio in the input composition is adjusted or altered compared to the ratio of the CCR7+CD45RA+CD4+ T cells to naive-like CD8+ T cells in the biological sample from the subject.
  • the provided methods comprise contacting the input composition with an agent comprising a nucleic acid molecule encoding a recombinant receptor under conditions to introduce the nucleic acid encoding the recombinant receptor into cells in the input composition.
  • the method comprises: contacting an input composition comprising CCR7+CD45RA+CD4+ T cells and CCR7+CD45RA+CD8+ T cells from a biological sample from a subject with an agent comprising a nucleic acid molecule encoding a recombinant receptor under conditions to introduce the nucleic acid encoding the recombinant receptor into cells in the composition, wherein the ratio of CCR7+CD45RA+CD4+ T cells to CCR7+CD45RA+CD8+ T cells present in the input composition is between or about between 0.8:1 and 2.2:1, inclusive.
  • stimulating the cells prior to, during and/or subsequent to said contacting, wherein stimulating comprises incubating the cells in the presence of one or more stimulating agents, wherein stimulating results in activation and/or proliferation of the cells.
  • the method comprises: combining a first cell composition comprising CCR7+CD45RA+CD4+ T cells with a second cell composition comprising CCR7+CD45RA+CD8+ T cells to produce an input cell composition in which the ratio of CCR7+CD45RA+CD4+ T cells to CCR7+CD45RA+CD8+ T cells is between or about between 0.8:1 and 2.2:1, inclusive; contacting the input composition with an agent comprising a nucleic acid molecule encoding a recombinant receptor under conditions to introduce the nucleic acid encoding the recombinant receptor into cells in the composition; and stimulating the cells, prior to, during and/or subsequent to said contacting, wherein stimulating comprises incubating the cells in the presence of one or more stimulating agents, wherein stimulating results in activation and/or proliferation of the cells.
  • the number, number per volume, number per weight, and/or percentage of CCR7+CD45RA+CD4+ T cells and/or the number, number per volume, number per weight, and/or percentage of CCR7+CD45RA+CD8+ T cells is determined by flow cytometry. In some embodiments of the provided methods, the ratio of CCR7+CD45RA+CD4+ T cells to CCR7+CD45RA+CD8+ T cells has been adjusted compared to the ratio of the CCR7+CD45RA+CD4+ T cells to CCR7+CD45RA+CD8+ T cells in a biological sample from the subject.
  • the input composition comprises a ratio of CCR7+CD45RA+CD4+ cells to CCR7+CD45RA+CD8+ cells of between or about between 0.8:1 and 2.0:1, 0.8:1 and 1.6:1, 0.8:1 and 1.4:1, 0.8:1 and 1.2:1, or 1.0:1 and 1.2:1, each inclusive.
  • the input composition comprises a ratio of CCR7+CD45RA+CD4+ cells to CCR7+CD45RA+CD8+ cells of or about 1.5:1, 1.4:1, 1.3:1, 1.2:1, 1.1:1, or 1.0:1.
  • the input composition comprises a ratio of CCR7+CD45RA+CD4+ cells to CCR7+CD45RA+CD8+ cells of or about 1.1:1.
  • the method comprises: combining a first cell composition comprising CD27+CCR7+CD4+ T cells with a second cell composition comprising CD27+CCR7+CD8+ T cells to produce an input cell composition in which the ratio of CD27+CCR7+CD4+ T cells to CD27+CCR7+CD8+ T cells is between or about between 0.8:1 and 2.2:1, inclusive.
  • the first cell composition is produced by isolating CD4+ T cells from a biological sample obtained from a subject and/or the second cell composition is produced by isolating CD8+ T cells from the biological sample obtained from the subject.
  • the method comprises determining the number, number per volume, number per weight, and/or percentage of the CD27+CCR7+CD4+ T cells in the first cell composition and/or the number, number per volume, number per weight, and/or percentage of the CD27+CCR7+CD8+ T cells in the second composition.
  • the method comprises determining the number, number per volume, number per weight, and/or percentage of the CD27+CCR7+CD4+ T cells and/or the number, number per volume, number per weight, and/or percentage of CD27+CCR7+CD8+ T cells in the biological sample from the subject.
  • the ratio of theCD27+CCR7+CD4+ T cells to CD27+CCR7+CD8+ T cells in the input composition is adjusted or altered compared to the ratio of the CD27+CCR7+CD4+ T cells to CD27+CCR7+CD8+ T cells in the biological sample from the subject.
  • a method for generating a cell composition comprising: determining the number, number per volume, number per weight, and/or percentage of CD27+CCR7+CD4+ T cells and CD27+CCR7+CD8+ T cells in a biological sample obtained from a subject or in one or more samples derived therefrom; and producing an input composition comprising CD4+ T cells and CD8+ T cells in which the ratio of the CD27+CCR7+CD4+ T cells to naive-like CD8+ T cells is between or about between 2.2:1 to 0.8:1, inclusive, wherein said ratio in the input composition is adjusted or altered compared to the ratio of the CD27+CCR7+CD4+ T cells to naive-like CD8+ T cells in the biological sample from the subject.
  • the provided methods comprise contacting the input composition with an agent comprising a nucleic acid molecule encoding a recombinant receptor under conditions to introduce the nucleic acid encoding the recombinant receptor into cells in the input composition.
  • the method comprises: contacting an input composition comprising CD27+CCR7+CD4+ T cells and CD27+CCR7+CD8+ T cells from a biological sample from a subject with an agent comprising a nucleic acid molecule encoding a recombinant receptor under conditions to introduce the nucleic acid encoding the recombinant receptor into cells in the composition, wherein the ratio of CD27+CCR7+CD4+ T cells to CD27+CCR7+CD8+ T cells present in the input composition is between or about between 0.8:1 and 2.2:1, inclusive.
  • Certain embodiments of the provided methods further comprise stimulating the cells, prior to, during and/or subsequent to said contacting, wherein stimulating comprises incubating the cells in the presence of one or more stimulating agents, wherein stimulating results in activation and/or proliferation of the cells.
  • the method comprises: combining a first cell composition comprising CD27+CCR7+CD4+ T cells with a second cell composition comprising CD27+CCR7+CD8+ T cells to produce an input cell composition in which the ratio of CD27+CCR7+CD4+ T cells to CD27+CCR7+CD8+ T cells is between or about between 0.8:1 and 2.2:1, inclusive; contacting the input composition with an agent comprising a nucleic acid molecule encoding a recombinant receptor under conditions to introduce the nucleic acid encoding the recombinant receptor into cells in the composition; and stimulating the cells, prior to, during and/or subsequent to said contacting, wherein stimulating comprises incubating the cells in the presence of one or more stimulating agents, wherein stimulating results in activation and/or proliferation of the cells.
  • the number, number per volume, number per weight, and/or percentage of CD27+CCR7+CD4+ T cells and/or the number, number per volume, number per weight, and/or percentage of CD27+CCR7+CD8+ T cells is determined by flow cytometry.
  • the ratio of CD27+CCR7+CD4+ T cells to CD27+CCR7+CD8+ T cells has been adjusted compared to the ratio of the CD27+CCR7+CD4+ T cells to CD27+CCR7+CD8+ T cells in a biological sample from the subject.
  • the input composition comprises a ratio of CD27+CCR7+CD4+ cells to CD27+CCR7+CD8+ cells of between or about between 0.8:1 and 2.0:1, 0.8:1 and 1.6:1, 0.8:1 and 1.4:1, 0.8:1 and 1.2:1, or 1.0:1 and 1.2:1, each inclusive.
  • the input composition comprises a ratio of CD27+CCR7+CD4+ cells to CD27+CCR7+CD8+ cells of or about 1.5:1, 1.4:1, 1.3:1, 1.2:1, 1.1:1, or 1.0:1.
  • the input composition comprises a ratio of CD27+CCR7+CD4+ cells to CD27+CCR7+CD8+ cells of or about 1.1:1. In some embodiments, the input cell composition comprises a ratio of CD27+CCR7+CD4+ cells to CD27+CCR7+CD8+ cells of or about 1.69:1.
  • the method comprises: combining a first cell composition comprising CD62L-CCR7+CD4+ T cells with a second cell composition comprising CD62L-CCR7+CD8+ T cells to produce an input cell composition in which the ratio of CD62L-CCR7+CD4+ T cells to CD62L-CCR7+CD8+ T cells is between or about between 0.8:1 and 2.2:1, inclusive.
  • the method comprises: determining the number, number per volume, number per weight, and/or percentage of CD62L-CCR7+CD4+ T cells and CD62L-CCR7+CD8+ T cells in a biological sample obtained from a subject or in one or more samples derived therefrom; and producing an input composition comprising CD4+ T cells and CD8+ T cells in which the ratio of the CD62L-CCR7+CD4+ T cells to naive-like CD8+ T cells is between or about between0.8:1 and 2.2:1, inclusive, wherein said ratio in the input composition is adjusted or altered compared to the ratio of the CD62L-CCR7+CD4+ T cells to naive-like CD8+ T cells in the biological sample from the subject.
  • the provided methods further comprise contacting the input composition with an agent comprising a nucleic acid molecule encoding a recombinant receptor under conditions to introduce the nucleic acid encoding the recombinant receptor into cells in the input composition.
  • the method comprises: combining a first cell composition comprising CD62L-CCR7+CD4+ T cells with a second cell composition comprising CD62L-CCR7+CD8+ T cells to produce an input cell composition in which the ratio of CD62L-CCR7+CD4+ T cells to CD62L-CCR7+CD8+ T cells is between or about between 0.8:1 and 2.2:1, inclusive; contacting the input composition with an agent comprising a nucleic acid molecule encoding a recombinant receptor under conditions to introduce the nucleic acid encoding the recombinant receptor into cells in the composition; and stimulating the cells, prior to, during and/or subsequent to said contacting, wherein stimulating comprises incubating the cells in the presence of one or more stimulating agents, wherein stimulating results in activation and/or proliferation of the cells.
  • the number, number per volume, number per weight, and/or percentage of CD62L-CCR7+CD4+ T cells and/or the number, number per volume, number per weight, and/or percentage of CD62L-CCR7+CD8+ T cells is determined by flow cytometry.
  • the ratio of CD62L-CCR7+CD4+ T cells to CD62L-CCR7+CD8+ T cells has been adjusted compared to the ratio of the CD62L-CCR7+CD4+ T cells to CD62L-CCR7+CD8+ T cells in a biological sample from the subject.
  • the input composition comprises a ratio of CD62L-CCR7+CD4+ cells to CD62L-CCR7+CD8+ cells of between or about between 1:1 and 2:1, or 0.8:1 and 1.2:1, each inclusive.
  • the input composition comprises a ratio of CD62L-CCR7+CD4+ cells to CD62L-CCR7+CD8+ cells of or about 1.2:1, 1.1:1, 1.0:1, 0.9:1, or 0.8:1.
  • the biological sample is or is obtained from a blood, plasma or serum sample.
  • the biological sample is or comprises a whole blood sample, a buffy coat sample, a peripheral blood mononuclear cells (PBMC) sample, an unfractionated T cell sample, a lymphocyte sample, a white blood cell sample, an apheresis product, or a leukapheresis product.
  • the biological sample is or is obtained from an apheresis or leukapheresis sample.
  • the methods include one or more steps for producing an input cell composition containing a defined, controlled, or desired ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells.
  • the disclosed input composition can be produced by mixing or combining a cell composition containing CD4+ T cells having a known or determined number or percentage of na ⁇ ve-like CD4+ T cells with a cell composition containing CD8+ T cells having a known or determined number or percentage of na ⁇ ve-like CD8+ T cells, such as to achieve the chosen or desired ratio.
  • the disclosed methods also can include methods associated with genetic engineering of cells, such as transduction methods, including methods for introducing a recombinant receptor, e.g. chimeric antigen receptor, into such cells for use in connection with adoptive cell therapy.
  • transduction methods including methods for introducing a recombinant receptor, e.g. chimeric antigen receptor, into such cells for use in connection with adoptive cell therapy.
  • processing of the generated input composition includes incubating the cells under stimulating conditions, for example in some aspects, to activate the cells for engineering or transduction or for cell expansion.
  • the methods include steps for engineering a plurality of cell types, such as CD4+ cells and CD8+ cells, such as those isolated and present in the input composition.
  • the engineering is carried out to introduce a genetically engineered antigen receptor into the cells, such as a TCR, e.g., a high-affinity TCR, or a functional non-TCR antigen receptor, such as a chimeric antigen receptor (CAR).
  • a TCR e.g., a high-affinity TCR
  • a functional non-TCR antigen receptor such as a chimeric antigen receptor (CAR).
  • the methods include further processing, such as further incubation, for example at or about 37° C ⁇ 2° C, and/or formulating of cells and compositions containing the same.
  • the processing produces a resulting output composition containing genetically engineered cells, such as genetically engineered CD4+ cells and CD8+ cells, including cells in which the engineered CD4+ and CD8+ cells are present at a desired ratio.
  • the resulting processed output composition can be used in methods for administering cells and compositions prepared by the methods to a patient, for example, in connection with adoptive cell therapy.
  • an output composition containing engineered cells e.g. CAR+ T cells, in which the engineered CD4+ cells and CD8+ cells are present at a desired ratio, or within a certain degree of tolerated error of the desired ratio, is advantageous.
  • engineered cells e.g. CAR+ T cells
  • CD4+ cells and CD8+ cells are present at a desired ratio, or within a certain degree of tolerated error of the desired ratio.
  • engineering cells enriched for a plurality of different cell populations or types of cells such as isolated CD4 + and CD8 + T cell populations and sub-populations, can improve efficacy of or reduce or avoid unwanted effects.
  • ratios include those deemed optimal for the therapeutic use, e.g., output ratios deemed appropriate or optimal for administration to a patient in connection with adoptive cell therapy.
  • the desired ratio of CD4+ to CD8+ T cells in an output composition for administration to a subject is from or from about 2:1 to 0.5:1, for example at or about 1:1.
  • a composition containing isolated CD8+ and CD4+ T cells such as containing a certain desired ratio of such cells, increases the ability of cells ultimately administered to a subject to persist, expand, become activated, and/or engraft in vivo or upon administration to a subject. In some aspects, it improves or increases one or more effector function or activation phenotype. For example, such advantages can be achieved in some aspects by administering a CD4+ and a CD8+ population in comparison with a CD8+ population alone.
  • the methods provide one or more advantages compared with other preparation, isolation, incubation, and engineering methods, such as cost, time, and/or resource savings.
  • advantages can include the ability to isolate, process, e.g., incubate, and/or engineer the plurality of cell populations, present at or near a desired ratio, with increased efficiency and/or reduced complexity, time, cost, and/or use of resources, compared with other methods.
  • the disclosed methods are based on observations that, in certain cell production processes for engineering cells, the input ratio of CD4+ to CD8+ cells, or a ratio of viable cells thereof, may not correlate to the output ratio of the engineered CD4+ to CD8+ cells, or a ratio of viable cells thereof. As shown herein, it is found that the desired ratio of engineered CD4+ to CD8+ T cells (e.g.
  • CAR+ CD4 to CAR+ CD8+ T cells or of the ratio of viable cells thereof) in an output composition is correlated or associated with the ratio of the na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells, or a ratio of viable cells thereof, present in an input composition prior to carrying out a production process on the cells to produce the output composition, e.g. involving one or more steps of stimulation, activation, expansion, proliferation and/or transduction of the cells.
  • Exemplary of such na ⁇ ve-like cells are ratios of CD4+ to CD8+ cells that are CD45RA+ and CCR7+, CD62L-/CCR7+ or CD27+/CCR7+.
  • the disclosed methods are based on observations that the desired ratio of engineered CD4+ to CD8+ T cells (e.g. CAR+ CD4 to CAR+ CD8+ T cells or of the ratio of viable cells thereof) in an output composition is correlated or associated with the ratio of the CD45RA+/CCR7+CD4+ T cells to CD45RA+/CCR7+CD8+ T cells, or a ratio of viable cells thereof, present in an input composition prior to carrying out a production process on the cells to produce the output composition, e.g. involving one or more steps of stimulation, activation, expansion, proliferation and/or transduction of the cells.
  • a production process e.g. involving one or more steps of stimulation, activation, expansion, proliferation and/or transduction of the cells.
  • CD45RA+/CCR7+CD4+ to CD45RA+/CCR7+/CD8+ cells in an input composition to engineered CD4+ to CD8+ cells (e.g. CAR+ CD4 to CAR+ CD8+ T cells or of the ratio of viable cells thereof) in an output composition exists even despite variations in the donor and/or process used to generate or produce the output compositions.
  • the disclosed methods are based on observations that the desired ratio of engineered CD4+ to CD8+ T cells (e.g. CAR+ CD4 to CAR+ CD8+ T cells or of the ratio of viable cells thereof) in an output composition is correlated or associated with the ratio of the CD62L-/CCR7+CD4+ T cells to CD62L-/CCR7+CD8+ T cells, or a ratio of viable cells thereof, present in an input composition prior to carrying out a production process on the cells to produce the output composition, e.g. involving one or more steps of stimulation, activation, expansion, proliferation and/or transduction of the cells.
  • a production process e.g. involving one or more steps of stimulation, activation, expansion, proliferation and/or transduction of the cells.
  • CD62L-/CCR7+CD4+ to CD62L-/CCR7+/CD8+ cells in an input composition to engineered CD4+ to CD8+ cells (e.g. CAR+ CD4 to CAR+ CD8+ T cells or of the ratio of viable cells thereof) in an output composition exists even despite variations in the donor and/or process used to generate or produce the output compositions.
  • the disclosed methods are based on observations that the desired ratio of engineered CD4+ to CD8+ T cells (e.g. CAR+ CD4 to CAR+ CD8+ T cells or of the ratio of viable cells thereof) in an output composition is correlated or associated with the ratio of the CD27+/CCR7+CD4+ T cells to CD27+/CCR7+CD8+ T cells, or a ratio of viable cells thereof, present in an input composition prior to carrying out a production process on the cells to produce the output composition, e.g. involving one or more steps of stimulation, activation, expansion, proliferation and/or transduction of the cells.
  • a production process e.g. involving one or more steps of stimulation, activation, expansion, proliferation and/or transduction of the cells.
  • the disclosed methods ensure that an output composition of genetically engineered cells, e.g. CAR+ T cells, generated by a cell production process achieves a relatively consistent and/or controlled desired ratio of the engineered CD4+ to CD8+ T cells, or a ratio of viable cells thereof, that exhibits a variance of such ratio that is low or below an acceptable or threshold variance, among compositions produced by the process, including those derived from samples from a number of different subjects, such as those having different characteristics, such as subjects of different ages, numbers and/or types of prior therapies, and indication and subtype or severity or grade thereof.
  • such processes generate a ratio of such engineered CD4+ to CD8+ (e.g.
  • CAR+ CD4 to CAR+ CD8+ T cells or of the ratio of viable cells thereof) that varies by no more than 20% or no more than 10% or no more than 5% from an average of said ratio in a plurality of T cell compositions produced by the process and/or varies from such average by no more than one standard deviation or varies by no more than 20% or no more than 10% or no more than 5% among a plurality of T cell compositions produced by the process among such various samples and patients.
  • the use of a process that yields greater consistency among the output composition generated among a cell production process, such as with respect to the engineered CD4 to CD8 T cell ratio, can be advantageous to ensure consistency in dosing of a subject, which, in some aspects, can optimize efficacy, potency and/or safety of the administered composition among treated subjects.
  • engineering cells enriched for CD4 + and CD8 + T cell populations at a desired output ratio in the engineered output composition can improve efficacy of or reduce or avoid unwanted effects.
  • the isolation or enrichment increases the ability of cells ultimately administered to a subject to persist, expand, become activated, and/or engraft in vivo or upon administration to a subject. In some aspects, it improves or increases one or more effector function or activation phenotype. Such results can be achieved even where there is donor variability among the starting cell samples for cell engineering.
  • the methods disclosed herein allow for the production of therapeutic cell compositions of engineered cells having a desired output ratio in the output composition without requiring the engineered CD4+ and CD8+ T cells to be processed and/or administered separately.
  • the disclosed methods provide for more streamlined and/or controlled methods for preparing a composition having at or near a desired output ratio of engineered CD4+ T cell population and CD8+ T cell population.
  • the disclosed methods can be used in a cell production process in which the CD4+ and CD8+ T cells are processed, e.g. activated, stimulated, expanded and/or transduced, together in a single stream process.
  • the methods permit for introduction of a genetically engineered antigen receptor for use in adoptive cell therapy, where the cell populations are isolated, incubated, and/or engineered in combination, and the method is associated with increased efficiency and/or reduced complexity, time, cost, and/or use of resources compared to a method in which the populations are isolated, incubated, and/or engineered separately.
  • cells and compositions prepared by the methods including pharmaceutical compositions and formulations, and kits, systems, and devices for carrying out the methods.
  • methods for use of the cells and compositions prepared by the methods including therapeutic methods, such as methods for adoptive cell therapy, and pharmaceutical compositions for administration to subjects.
  • the input cell composition contains CD4+ T cells and CD8+ T cells.
  • the input cell composition one or more subtypes or populations of CD4+ and/or CD8+ T cells.
  • the one or more subtypes or populations are naive and/or na ⁇ ve-like cells.
  • the input cell composition contains CD4+ T cells, and at least a portion of the CD4+ T cells are na ⁇ ve-like CD4 cells.
  • the input cell composition contains CD8+ T cells, and at least a portion of the CD8+ T cells are naive cells.
  • the input cell composition contains and/or has a fixed, preferred, target, defined, and/or controlled ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells.
  • the methods include one or more steps of mixing or combining cells or compositions of cells to generate or produce an input cell composition.
  • the cells or cell compositions have been selected and/or isolated from a sample.
  • the cells of the input cell composition have been selected and/or isolated from a sample.
  • CD4+ T cells and/or a composition of CD4+ T cells are selected or isolated from a sample.
  • the sample is a biological sample, such as blood sample, apheresis sample, and/or leukapheresis sample.
  • the sample is from a subject, e.g. a human subject.
  • composition of CD4+ T cells and the composition of CD8+ T cells are isolated and/or selected from the same sample.
  • the composition of CD4+ T cells and the composition of CD8+ T cells are isolated and/or selected from samples taken or obtained from the same subject.
  • the generation or production of an input cell composition includes one or more steps of assessing, characterizing, and/or identifying cells.
  • the cells are assessed, characterized, and/or identified in a composition of CD4+ T cells.
  • cells are assessed, characterized, and/or identified in a composition of CD8+ T cells.
  • the cells are assessed for cells that are positive for a marker that indicates and/or is associated with a na ⁇ ve-like state in T cells.
  • the cells are assessed for cells that are negative for a marker that indicates and/or is associated with a non-na ⁇ ve-like state in T cells.
  • the cells from CD4+ and CD8+ T cell compositions are assessed, characterized, and/or identified to determine the amount, level, portion, and/or percentage of cells that are positive for one or more markers associated with a na ⁇ ve-like state and/or are negative for one or more markers associated with a non-na ⁇ ve-like state.
  • the cells from CD4+ and CD8+ T cell compositions such as cell compositions isolated from a biological sample from a subject, are assessed, characterized, and/or identified to determine the amount, level, portion, and/or percentage of cells that are na ⁇ ve-like cells.
  • the methods include one or more steps of mixing or combining a cell composition containing CD4+ T cells with a cell composition containing CD8+ T cells to generate or produce a cell composition, e.g., an input cell composition, with a defined ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells.
  • the generation or production of the input cell composition includes one or more steps of: (i) isolating or selecting a composition of CD4+ T cells and/or a composition of CD8+ T cells from a sample, e.g., a biological sample; (ii) assessing, characterizing, and/or identifying the amount, level, portion, and/or percentage of cells that are positive for one or more markers associated with a na ⁇ ve-like state and/or are negative for one or more markers associated with a non-na ⁇ ve-like state in the composition of CD4+ and/or CD8+ T cells; and/or (iii) mixing or combining cells of a composition of CD4+ T cells with cells of a composition of CD8+ T cells at a defined, fixed, and/or preferred ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells.
  • the content, make-up, and/or constitution of the input cell composition correlates, controls, corresponds, and/or associates with the content, make-up, and/or constitution of the output cell composition.
  • the amount, portion, percentage, number, number per volume, number per weight, and/or ratio of na ⁇ ve and/or na ⁇ ve-like cells, e.g., na ⁇ ve-like T cells, in an input cell composition correlates, controls, corresponds, and/or associates with the with the content, make-up, and/or constitution of the output cell composition.
  • the amount, portion, number, number per volume, number per weight, and/or ratio of na ⁇ ve-like CD4+ T cells and/or na ⁇ ve-like CD8+ T cells in an input cell composition correlates, controls, corresponds, and/or associates with the with the content, make-up, and/or constitution of the output cell composition.
  • the methods include one or more steps of genetically engineering the cells of the input composition.
  • the genetic engineering includes one or more steps of incubating the cells of the input cell composition under conditions that activate and/or simulate the cells, delivering a gene, e.g., a recombinant and/or heterologous gene to the cells, expanding the cells by incubating the cells under activing or stimulating conditions, harvesting the cells, and/or storing the cells by freezing, e.g., cryopreservation.
  • the one or more steps of genetic engineering produce an output cell composition containing engineered cells.
  • engineered cells of the output composition have a fixed, defined, and/or target ratio of CD4+ to CD8+ cells.
  • the methods disclosed herein include one or more steps for generating engineered cells, such as cells expressing a recombinant receptor, that have a defined ratio of CD4+ to CD8+ T cells.
  • the methods disclosed herein include one or more steps for genetically engineering cells from a starting and/or input cell composition to generate a resulting and/or an output cell composition having a defined ratio genetically engineered CD4+ to CD8+ T cells.
  • the genetic engineering is or includes transfection or transduction of cells from the input cell composition to introduce an agent containing a nucleic acid into the cells of the input cell composition.
  • the nucleic acid encodes a recombinant receptor, e.g., a chimeric antigen receptor (CAR).
  • generating an output cell composition includes one or more steps of activating or stimulating cells of the input cell composition; genetically engineering, transducing or transfecting the cells from the input cell composition; and/or expanding the transfected cells; thereby resulting in output cell composition having a defined ratio genetically engineered CD4+ to CD8+ T cells, such as a 1:1 ratio.
  • the methods disclosed herein include one or more steps for preparing cells for genetic engineering.
  • the one or more steps include isolating cells from a biological sample to prepare a composition of cells to be genetically engineered, e.g., an input cell composition.
  • the preparation of an input cell composition includes one or more steps of isolating two or more cell types and/or compositions of a particular cell type or cell subtype, and mixing or combining the cell types and/or the cell compositions of the particular cell types into a single input cell composition.
  • the cell compositions of the particular cell types are assessed to determine the presence, amount, and/or ratios of further subtypes in the compositions.
  • the cell compositions of the particular cell types are mixed or combined to achieve input cell compositions of at fixed or determined ratios of cell types or subtypes.
  • the cell types and/or cell subtypes correlate, control, correspond, and/or associate with the content, make-up, and/or constitution of the output cell composition.
  • the preparation of an input cell composition includes one or more steps of isolating compositions of or including CD4+ T cells, isolating compositions of or including CD8+ T cells, and mixing or combining the CD4+ and CD8+ T cell compositions of the particular cell types into a single input cell composition.
  • the preparation of the input cell composition includes one or more steps of assessing, determining, and/or quantifying the portion or amount of a subtype of the CD4+ and/or CD8+ T cells of the cell compositions.
  • the subtype of the CD4+ and/or CD8+ T cells are or include naive and/or na ⁇ ve-like CD4+ and/or CD8+ T cells.
  • the portions or amounts of na ⁇ ve-like cells in the input cell composition correlates, controls, corresponds, and/or associates with the content, make-up, and/or constitution of the output cell composition.
  • the cells e.g., the CD4+ and/or CD8+ T cells, that are isolated from a sample are eukaryotic cells, such as mammalian cells, and in some instances are human cells.
  • the cells are derived from the blood, bone marrow, lymph, or lymphoid organs of a subject, and/or are cells of the immune system, such as cells of the innate or adaptive immunity.
  • the cells are lymphocytes.
  • the lymphocytes are T lymphocytes or T cells.
  • the cells include CD4+ T cells and CD8+ T cells.
  • compositions of T cells contain subtypes of cells that are further categorized by function, activation state, maturity, potential for differentiation, expansion, marker or cytokine secretion profile, and/or degree of differentiation.
  • the cells are or include naive and/or na ⁇ ve-like CD4+ and/or CD8+ T cells.
  • the cells may be allogeneic and/or autologous.
  • the methods include isolating cells from the subject, preparing, processing, culturing, and/or engineering the cells, and re-introducing the cells into the same subject, before or after cryopreservation.
  • preparation of the engineered cells includes one or more culture and/or preparation steps.
  • the cells for use in an input cell composition e.g., a composition of cells to be genetically engineered, e.g., to express a recombinant receptor such as the CAR, may contain cells that have been isolated from a sample, such as a biological sample, e.g., one obtained from or derived from a subject.
  • the subject from which the cell is isolated is one having the disease or condition or in need of a cell therapy or to which cell therapy will be administered.
  • the subject in some instances is a human in need of a particular therapeutic intervention, such as the adoptive cell therapy for which cells are being isolated, processed, and/or engineered.
  • the sample e.g., a biological sample, from which the cells are derived or isolated is blood or a blood-derived sample, or is derived from an apheresis or leukapheresis product.
  • exemplary samples include whole blood, peripheral blood mononuclear cells (PBMCs), leukocytes, bone marrow, thymus, and/or cells derived therefrom.
  • cells are derived, isolated, and/or selected from samples or biological samples that may include a whole blood sample, a buffy coat sample, a peripheral blood mononuclear cells (PBMC) sample, an unfractionated T cell sample, a lymphocyte sample, a white blood cell sample, an apheresis product, or a leukapheresis product.
  • Samples include, in the context of cell therapy, e.g., adoptive cell therapy, samples from autologous and allogeneic sources.
  • the cells are derived from cell lines, e.g., T cell lines.
  • the cells in some instances are obtained from a xenogeneic source, for example, from mouse, rat, non-human primate, and pig.
  • isolation of the cells or the compositions of cells includes one or more preparation and/or non-affinity based cell separation steps.
  • cells are washed, centrifuged, and/or incubated in the presence of one or more reagents, for example, to remove unwanted components, enrich for desired components, lyse or remove cells sensitive to particular reagents.
  • reagents for example, to remove unwanted components, enrich for desired components, lyse or remove cells sensitive to particular reagents.
  • cells are separated based on one or more property, such as density, adherent properties, size, sensitivity and/or resistance to particular components.
  • cells from the circulating blood of a subject are obtained, e.g., by apheresis or leukapheresis.
  • the samples contain lymphocytes, including T cells, monocytes, granulocytes, B cells, other nucleated white blood cells, red blood cells, and/or platelets, and in some aspects contains cells other than red blood cells and platelets.
  • the blood cells collected from the subject are washed, e.g., to remove the plasma fraction and to place the cells in an appropriate buffer or media for subsequent processing steps.
  • the cells are washed with phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the wash solution lacks calcium and/or magnesium and/or many or all divalent cations.
  • a washing step is accomplished a semi-automated "flow-through" centrifuge (for example, the Cobe 2991 cell processor, Baxter) according to the manufacturer's instructions.
  • a washing step is accomplished by tangential flow filtration (TFF) according to the manufacturer's instructions.
  • the cells are resuspended in a variety of biocompatible buffers after washing, such as, for example, Ca ++ /Mg ++ free PBS.
  • components of a blood cell sample are removed and the cells directly resuspended in culture media.
  • the methods include density-based cell separation methods, such as the preparation of white blood cells from peripheral blood by lysing the red blood cells and centrifugation through a Percoll or Ficoll gradient.
  • the isolation methods include the separation of different cell types based on the expression or presence in the cell of one or more specific molecules, such as surface markers, e.g., surface proteins, intracellular markers, or nucleic acid. In some instances, any known method for separation based on such markers may be used. In some instances, the separation is affinity- or immunoaffinity-based separation.
  • the isolation in some aspects includes separation of cells and cell populations based on the cells' expression or expression level of one or more markers, typically cell surface markers, for example, by incubation with an antibody or binding partner that specifically binds to such markers, followed generally by washing steps and separation of cells having bound the antibody or binding partner, from those cells having not bound to the antibody or binding partner.
  • Such separation steps can be based on positive selection, in which the cells having bound the reagents are retained for further use, and/or negative selection, in which the cells having not bound to the antibody or binding partner are retained. In some examples, both fractions are retained for further use. In some aspects, negative selection can be particularly useful where no antibody is available that specifically identifies a cell type in a heterogeneous population, such that separation is best carried out based on markers expressed by cells other than the desired population.
  • the separation need not result in 100% enrichment or removal of a particular cell population or cells expressing a particular marker.
  • positive selection of or enrichment for cells of a particular type refers to increasing the number or percentage of such cells, but need not result in a complete absence of cells not expressing the marker.
  • negative selection, removal, or depletion of cells of a particular type refers to decreasing the number or percentage of such cells, but need not result in a complete removal of all such cells.
  • multiple rounds of separation steps are carried out, where the positively or negatively selected fraction from one step is subjected to another separation step, such as a subsequent positive or negative selection.
  • a single separation step can deplete cells expressing multiple markers simultaneously, such as by incubating cells with a plurality of antibodies or binding partners, each specific for a marker targeted for negative selection.
  • multiple cell types can simultaneously be positively selected by incubating cells with a plurality of antibodies or binding partners expressed on the various cell types.
  • specific subpopulations or subtypes of T cells are isolated by positive or negative selection techniques.
  • CD4+ T cells and/or a cell composition of or including CD4+ T cells are isolated by positive or negative selection techniques.
  • CD8+ T cells and/or a cell composition of or including CD8+ T cells are isolated by positive or negative selection techniques.
  • a composition of CD4+ T cells and/or a composition of CD8+ T cells are isolated by positive or negative selection techniques.
  • isolation is carried out by enrichment for a particular cell population by positive selection, or depletion of a particular cell population, by negative selection.
  • positive or negative selection is accomplished by incubating cells with one or more antibodies or other binding agent that specifically bind to one or more surface markers expressed (marker + or marker+) or expressed at a relatively higher level (marker high ) on the positively or negatively selected cells, respectively.
  • T cells are separated from a PBMC sample by negative selection of markers expressed on non-T cells, such as B cells, monocytes, or other white blood cells, such as CD14.
  • a CD4 + or CD8 + selection step is used to separate CD4 + helper and CD8 + cytotoxic T cells.
  • Such CD4 + and CD8 + compositions can contain cells that can be further classified or sorted into sub-populations based on the positive or negative expression of markers and/or the relative level of expression of the markers.
  • subtypes may include naive, na ⁇ ve-like, and/or non-na ⁇ ve subtypes or subpopulations.
  • CD4+ T cells e.g., CD4+ T helper cells
  • CD4+ T helper cells are categorized into naive and/or na ⁇ ve-like, and non-naive and/or non-na ⁇ ve-like cells by identifying cell populations that have cell surface antigens.
  • CD8+ T cells e.g., CD8+ T helper cells
  • CD8+ T helper cells are categorized into naive and/or na ⁇ ve-like, and non-naive and/or non-na ⁇ ve-like cells by identifying cell populations that have cell surface antigens.
  • the T lymphocytes are separated or isolated using immunomagnetic (or affinitymagnetic) separation techniques.
  • CD4+ T cells and/or compositions of CD4+ T cells are separated or isolated using immunomagnetic separation techniques.
  • CD8+ T cells and/or compositions of CD8+ T cells are separated or isolated using immunomagnetic separation techniques. Separation and isolation using immunomagnetic (or affinitymagnetic) separation techniques are reviewed in Methods in Molecular Medicine, vol. 58 : Metastasis Research Protocols, Vol. 2 : Cell Behavior In Vitro and In Vivo, p 17-25 Edited by: S. A. Brooks and U. Schumacher ⁇ Humana Press Inc., Totowa, NJ .
  • the sample or composition of cells to be separated is incubated with small, magnetizable or magnetically responsive material, such as magnetically responsive particles or microparticles, such as paramagnetic beads (e.g., such as Dynalbeads or MACS beads).
  • the magnetically responsive material, e.g., particle generally is directly or indirectly attached to a binding partner, e.g., an antibody, that specifically binds to a molecule, e.g., surface marker, present on the cell, cells, or population of cells that it is desired to separate, e.g., that it is desired to negatively or positively select.
  • the magnetic particle or bead comprises a magnetically responsive material bound to a specific binding member, such as an antibody or other binding partner.
  • a magnetically responsive material used in magnetic separation methods. Suitable magnetic particles include those described in Molday, U.S. Pat. No. 4,452,773 , and in European Patent Specification EP 452342 B . Colloidal sized particles, such as those described in Owen U.S. Pat. No. 4,795,698 , and Liberti et al., U.S. Pat. No. 5,200,084 are other examples.
  • the incubation generally is carried out under conditions whereby the antibodies or binding partners, or molecules, such as secondary antibodies or other reagents, which specifically bind to such antibodies or binding partners, which are attached to the magnetic particle or bead, specifically bind to cell surface molecules if present on cells within the sample.
  • the antibodies or binding partners, or molecules, such as secondary antibodies or other reagents which specifically bind to such antibodies or binding partners, which are attached to the magnetic particle or bead, specifically bind to cell surface molecules if present on cells within the sample.
  • the sample is placed in a magnetic field, and those cells having magnetically responsive or magnetizable particles attached thereto will be attracted to the magnet and separated from the unlabeled cells.
  • positive selection cells that are attracted to the magnet are retained; for negative selection, cells that are not attracted (unlabeled cells) are retained.
  • negative selection cells that are not attracted (unlabeled cells) are retained.
  • a combination of positive and negative selection is performed during the same selection step, where the positive and negative fractions are retained and further processed or subject to further separation steps.
  • the magnetically responsive particles are coated in primary antibodies or other binding partners, secondary antibodies, lectins, enzymes, or streptavidin.
  • the magnetic particles are attached to cells via a coating of primary antibodies specific for one or more markers.
  • the cells, rather than the beads are labeled with a primary antibody or binding partner, and then cell-type specific secondary antibody- or other binding partner (e.g., streptavidin)-coated magnetic particles, are added.
  • streptavidin-coated magnetic particles are used in conjunction with biotinylated primary or secondary antibodies.
  • the magnetically responsive particles are left attached to the cells that are to be subsequently incubated, cultured and/or engineered; in some aspects, the particles are left attached to the cells for administration to a patient.
  • the magnetizable or magnetically responsive particles are removed from the cells. Methods for removing magnetizable particles from cells are known and include, e.g., the use of competing non-labeled antibodies, and magnetizable particles or antibodies conjugated to cleavable linkers. In some instances, the magnetizable particles are biodegradable.
  • the affinity-based selection is via magnetic-activated cell sorting (MACS) (Miltenyi Biotec, Auburn, CA). Magnetic Activated Cell Sorting (MACS) systems are capable of high-purity selection of cells having magnetized particles attached thereto.
  • MACS operates in a mode wherein the non-target and target species are sequentially eluted after the application of the external magnetic field. That is, the cells attached to magnetized particles are held in place while the unattached species are eluted. Then, after this first elution step is completed, the species that were trapped in the magnetic field and were prevented from being eluted are freed in some manner such that they can be eluted and recovered.
  • the non-target cells are labelled and depleted from the heterogeneous population of cells.
  • the isolation or separation is carried out using a system, device, or apparatus that carries out one or more of the isolation, cell preparation, separation, processing, incubation, culture, and/or formulation steps of the methods.
  • the system is used to carry out each of these steps in a closed or sterile environment, for example, to minimize error, user handling and/or contamination.
  • the system is a system as described in International Patent Application, Publication Number WO2009/072003 , or US 20110003380 A1 .
  • the system or apparatus carries out one or more, e.g., all, of the isolation, processing, engineering, and formulation steps in an integrated or self-contained system, and/or in an automated or programmable fashion.
  • the system or apparatus includes a computer and/or computer program in communication with the system or apparatus, which allows a user to program, control, assess the outcome of, and/or adjust various aspects of the processing, isolation, engineering, and formulation steps.
  • the separation and/or other steps is carried out using CliniMACS system (Miltenyi Biotec), for example, for automated separation of cells on a clinical-scale level in a closed and sterile system.
  • Components can include an integrated microcomputer, magnetic separation unit, peristaltic pump, and various pinch valves.
  • the integrated computer in some aspects controls all components of the instrument and directs the system to perform repeated procedures in a standardized sequence.
  • the magnetic separation unit in some aspects includes a movable permanent magnet and a holder for the selection column.
  • the peristaltic pump controls the flow rate throughout the tubing set and, together with the pinch valves, ensures the controlled flow of buffer through the system and continual suspension of cells.
  • the CliniMACS system in some aspects uses antibody-coupled magnetizable particles that are supplied in a sterile, non-pyrogenic solution.
  • the cells after labelling of cells with magnetic particles the cells are washed to remove excess particles.
  • a cell preparation bag is then connected to the tubing set, which in turn is connected to a bag containing buffer and a cell collection bag.
  • the tubing set consists of pre-assembled sterile tubing, including a pre-column and a separation column, and are for single use only. After initiation of the separation program, the system automatically applies the cell sample onto the separation column. Labelled cells are retained within the column, while unlabeled cells are removed by a series of washing steps.
  • the cell populations for use with the methods described herein are unlabeled and are not retained in the column. In some instances, the cell populations for use with the methods described herein are labeled and are retained in the column. In some instances, the cell populations for use with the methods described herein are eluted from the column after removal of the magnetic field, and are collected within the cell collection bag.
  • the CliniMACS Prodigy system in some aspects is equipped with a cell processing unity that permits automated washing and fractionation of cells by centrifugation.
  • the CliniMACS Prodigy system can also include an onboard camera and image recognition software that determines the optimal cell fractionation endpoint by discerning the macroscopic layers of the source cell product. For example, peripheral blood is automatically separated into erythrocytes, white blood cells and plasma layers.
  • the CliniMACS Prodigy system can also include an integrated cell cultivation chamber which accomplishes cell culture protocols such as, e.g., cell differentiation and expansion, antigen loading, and long-term cell culture.
  • Input ports can allow for the sterile removal and replenishment of media and cells can be monitored using an integrated microscope. See, e.g., Klebanoff et al. (2012) J Immunother. 35(9): 651-660 , Terakura et al. (2012) Blood.1:72-82 , and Wang et al. (2012) J Immunother. 35(9):689-701 .
  • a cell population described herein is collected and enriched (or depleted) via flow cytometry, in which cells stained for multiple cell surface markers are carried in a fluidic stream.
  • a cell population described herein is collected and enriched (or depleted) via preparative scale (FACS)-sorting.
  • FACS preparative scale
  • a cell population described herein is collected and enriched (or depleted) by use of microelectromechanical systems (MEMS) chips in combination with a FACS-based detection system (see, e.g., WO 2010/033140 , Cho et al. (2010) Lab Chip 10, 1567-1573 ; and Godin et al. (2008) J Biophoton. 1(5):355-376 . In both cases, cells can be labeled with multiple markers, allowing for the isolation of well-defined T cell subsets at high purity.
  • MEMS microelectromechanical systems
  • the antibodies or binding partners are labeled with one or more detectable marker, to facilitate separation for positive and/or negative selection.
  • separation may be based on binding to fluorescently labeled antibodies.
  • separation of cells based on binding of antibodies or other binding partners specific for one or more cell surface markers are carried in a fluidic stream, such as by fluorescence-activated cell sorting (FACS), including preparative scale (FACS) and/or microelectromechanical systems (MEMS) chips, e.g., in combination with a flow-cytometric detection system.
  • FACS fluorescence-activated cell sorting
  • MEMS microelectromechanical systems
  • the input cell composition is a composition of cells for use in genetic engineering, e.g., cells that will be genetically engineered with a nucleic acid encoding a recombinant protein, such as a recombinant receptor, e.g. CAR and/or that will undergo a process to produce genetically engineered cells expressing a recombinant protein, such as a recombinant receptor, e.g. a CAR.
  • the cells of the input composition will be treated with, contacted with, and/or incubated with a nucleic acid that encodes a recombinant receptor.
  • the input cell composition contains CD4+ T cells and CD8+ T cells.
  • the input cell composition contain CD4+ T cells and CD8+ T cells that include a particular desired, fixed and/or controlled ratio of CD4+ to CD8+ T cells that are naive and/or na ⁇ ve-like T cells.
  • the desired, fixed, and/or controlled ratio of CD4+ to CD8+ T cells that are naive and/or na ⁇ ve-like T cells is the ratio or number of cells at which two types of cells or isolated cell populations are included in an input cell composition, designed to result in an output cell composition with a desired, defined, and/or controlled ratio of engineered CD4+ to CD8+ T cells, or within a tolerated error rate or difference thereof, at the completion of the incubation and/or engineering step or other processing steps and/or upon thaw and/or just prior to administration to a subject.
  • the input cell composition contains a ratio, e.g., a defined, controlled, and/or fixed ratio, CD4+ na ⁇ ve-like T cells to CD8+ naive-like T cells.
  • the ratio of CD4+ na ⁇ ve-like T cells to CD8+ na ⁇ ve-like T cells is between 10:1 to 0.05:1, between 8:1 to 0.1:1, between 5:1 to 0.2:1, between 2.5:1 to 0.25:1, between 2.2:1 to 0.8:1, between 2:1 to 0.5:1, or between 1.5:1 to 1:1, inclusive.
  • the ratio of CD4+ na ⁇ ve-like T cells to CD8+ na ⁇ ve-like T cells is between 2:1 to 0.8:1, between 1.6:1 to 0.8:1, between 1.4:1 to 0.8:1, between 1.2:1 to 0.8:1,or between 1.2:1 to 0.8:1, inclusive. In some instances, the ratio is between 2.2:1 to 0.8:1, inclusive.
  • the ratio of CD4+ na ⁇ ve-like T cells to CD8+ na ⁇ ve-like T cells is or is about 2.2:1, 2.1:1, 2.0:1, 1.9:1, 1.8:1, 1.7:1, 1.6:1, 1.5:1, 1.4:1, 1.3:1, 1.2:1, 1.1:1, 1.0:1, 0.9:1, or 0.8:1. In certain instances, the ratio is or is about 1.1:1.
  • the input cell composition has an amount of or about of 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1.0 ⁇ 10 8 , 1.1 ⁇ 10 8 , 1.2 ⁇ 10 8 , 1.3 ⁇ 10 8 , 1.4 ⁇ 10 8 , 1.5 ⁇ 10 8 , 1.6 ⁇ 10 8 , 1.7 ⁇ 10 8 , 1.8 ⁇ 10 8 , 1.9 ⁇ 10 8 , 2.0 ⁇ 10 8 , 2.1 ⁇ 10 8 , 2.2 ⁇ 10 8 , 2.3 ⁇ 10 8 , 2.4 ⁇ 10 8 , 2.5 ⁇ 10 8 , 2.6 ⁇ 10 8 , 2.7 ⁇ 10 8 , 2.8 ⁇ 10 8 , 2.9 ⁇ 10 8 , 3.0 ⁇ 10 8 , 3.5 ⁇ 10 8 , 4.0 ⁇ 10 8 , 4.5 ⁇ 10 8 , 5 ⁇ 10 8 , 5 ⁇ 10 8 , or 1 ⁇ 10 9 total cells or
  • the input cell composition has an amount of or about of 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1.0 ⁇ 10 8 , 1.1 ⁇ 10 8 , 1.2 ⁇ 10 8 , 1.3 ⁇ 10 8 , 1.4 ⁇ 10 8 , 1.5 ⁇ 10 8 , 1.6 ⁇ 10 8 , 1.7 ⁇ 10 8 , 1.8 ⁇ 10 8 , 1.9 ⁇ 10 8 , 2.0 ⁇ 10 8 , 2.1 ⁇ 10 8 , 2.2 ⁇ 10 8 , 2.3 ⁇ 10 8 , 2.4 ⁇ 10 8 , 2.5 ⁇ 10 8 , 2.6 ⁇ 10 8 , 2.7 ⁇ 10 8 , 2.8 ⁇ 10 8 , 2.9 ⁇ 10 8 , 3.0 ⁇ 10 8 , 3.5 ⁇ 10 8 , 4.0 ⁇ 10 8 , 4.5 ⁇ 10 8 , 5 ⁇ 10 8 , 5 ⁇ 10 8 , or 1 ⁇ 10 9 cells that express
  • the input cell composition has an amount of or about of 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1.0 ⁇ 10 8 , 1.1 ⁇ 10 8 , 1.2 ⁇ 10 8 , 1.3 ⁇ 10 8 , 1.4 ⁇ 10 8 , 1.5 ⁇ 10 8 , 1.6 ⁇ 10 8 , 1.7 ⁇ 10 8 , 1.8 ⁇ 10 8 , 1.9 ⁇ 10 8 , 2.0 ⁇ 10 8 , 2.1 ⁇ 10 8 , 2.2 ⁇ 10 8 , 2.3 ⁇ 10 8 , 2.4 ⁇ 10 8 , 2.5 ⁇ 10 8 , 2.6 ⁇ 10 8 , 2.7 ⁇ 10 8 , 2.8 ⁇ 10 8 , 2.9 ⁇ 10 8 , 3.0 ⁇ 10 8 , 3.5 ⁇ 10 8 , 4.0 ⁇ 10 8 , 4.5 ⁇ 10 8 , 5 ⁇ 10 8 , 5 ⁇ 10 8 , or 1 ⁇ 10 9 na
  • the input cell composition has between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total cells or total viable cells. In certain instances, the input cell composition has an amount of or about of between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 cells that express CD4 or CD8.
  • the input cell composition has an amount of or about of between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 na ⁇ ve-like CD4+ and na ⁇ ve-like CD8+ T cells.
  • the input cell composition has or contains at least 1%, at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70% , at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or 100% or about 100% na ⁇ ve-like cells.
  • the input cell composition contains or includes no more than 100%, no more than 99%, no more than 98%, no more than 97%, no more than 96%, no more than 95%, no more than 90%, or no more than 85% na ⁇ ve-like cells.
  • the methods disclosed herein include one or more steps of producing, generating, and/or making an input cell composition.
  • the producing, generating, and/or making an input cell composition includes one or more steps of mixing or combining a cells of a composition of CD4+ T cells with cells of a composition of CD8+ T cells.
  • the cells, e.g., the CD4+ T cells CD8+ T cells, of the input composition have been isolated and/or selected from a sample, e.g., a biological sample.
  • the source of the cells of the input composition are compositions of cells, e.g., compositions of CD4+ and compositions of CD8+ T cells, that have been isolated and/or selected from the sample, such as separately isolated or selected.
  • the composition of CD4+ T cells and the composition of CD8+ T cells are isolated and/or selected, such as separately isolated and/or selected, from a sample, e.g., a biological sample.
  • composition of CD4+ T cells and the composition of CD8+ T cells are isolated and/or selected from the same sample. In certain instances, the composition of CD4+ T cells and the composition of CD8+ T cells are isolated and/or selected from samples taken or obtained from the same subject.
  • the composition of CD4+ T cells contains or includes at least 60%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or 100% or about 100% CD4+ T cells.
  • the composition of CD4+ T cells contains or includes no more than 100%, no more than 99%, no more than 98%, no more than 97%, no more than 96%, no more than 95%, no more than 90%, or no more than 85% CD4+ T cells.
  • the composition of CD8+ T cells contains or includes at least 60%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or 100% or about 100% CD8+ T cells.
  • the composition of CD8+ T cells contains or includes no more than 100%, no more than 99%, no more than 98%, no more than 97%, no more than 96%, no more than 95%, no more than 90%, or no more than 85% CD8+ T cells.
  • the producing, generating, and/or making an input cell composition includes one or more steps of measuring, determining, and/or quantifying the amount, portion, number, number per volume, number per weight, and/or percentage of viable CD4+ T cells and/or viable CD8+ T cells that are present in the composition of CD4+ T cells and/or the composition of CD8+ T cells, e.g., prior to combining or mixing the cells of the cell compositions.
  • the producing, generating, and/or making an input cell composition includes one or more steps of measuring, determining, and/or quantifying the amount, portion, number, number per volume, number per weight, and/or percentage of na ⁇ ve-like CD4+ T cells and/or na ⁇ ve-like CD8+ T cells that are present in the composition of CD4+ T cells and/or the composition of CD8+ T cells.
  • the na ⁇ ve-like CD4+ and/or na ⁇ ve-like CD8+ T cells are viable na ⁇ ve-like cells.
  • the producing, generating, and/or making an input cell composition includes one or more steps of measuring, determining, and/or quantifying the amount, portion, number, number per volume, number per weight, and/or percentage of viable CD4+ T cells and/or viable CD8+ T cells that are present in the sample, e.g., the biological sample.
  • the producing, generating, and/or making an input cell composition includes one or more steps of measuring, determining, and/or quantifying the amount, portion, number, number per volume, number per weight, and/or percentage of na ⁇ ve-like CD4+ T cells and/or na ⁇ ve-like CD8+ T cells that are present in the sample.
  • the na ⁇ ve-like CD4+ and/or na ⁇ ve-like CD8+ T cells are viable na ⁇ ve-like cells.
  • the cells of the input composition are isolated and/or selected from a sample, e.g., a biological sample.
  • a sample e.g., a biological sample.
  • the portions of na ⁇ ve-like cells in the sample e.g., portion of na ⁇ ve-like CD4+ and CD8+ T cells are known or have been determined, measured, or assessed.
  • cells from the sample are isolated and/or selected to directly produce a cell composition, e.g., an input cell composition, with a defined, fixed, or controlled ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells.
  • the cells are isolated and/or selected with immunoaffinity bead selection.
  • the cells are isolated and/or selected with affinity columns.
  • the cells from the sample are isolated or selected according to any of the methods described in WO 2015/164675 to produce a cell composition with a defined, controlled, and/or fixed ratio of na ⁇ ve-like CD4+ cells to na ⁇ ve-like CD8+ cells.
  • the input cell composition contains cells that were directly isolated and/or selected from a sample by a first and second isolation or selection.
  • the input composition is produced by performing a first and second selection to isolate an amount, number, or concentration of CD4+ T cells and CD8+ T cells sufficient to produce the defined, fixed, and/or controlled ratio of na ⁇ ve-like CD4+ to na ⁇ ve-like CD8+ T cells.
  • the cells from the sample are directly isolated, selected, and/or enriched to produce an input cell composition enriched for CD4+ cells and CD8+ cells.
  • the amount, number, percentage, number per volume, and/or number per weight of na ⁇ ve-like CD4+ and na ⁇ ve-like CD8+ cells have been measured, assessed, and/or determined in the sample, and the CD4+ and CD8+ cells are isolated, selected, and/or enriched in sufficient amounts to achieve an input cell composition with the defined, fixed, or controlled ratio of na ⁇ ve-like CD4+ to na ⁇ ve-like CD8+ T cells.
  • the cells that are directly isolated, selected, and/or enriched from the sample are the input cell composition and are used in subsequent processing steps, such as subsequent processing steps involving incubation, stimulation, activation, engineering and/or formulation of the enriched cells.
  • the isolated, selected, and/or enriched cells from the sample such as an input cell composition, contain a ratio of CD4+ cells to CD8+ cells at a defined, fixed, or controlled ratio of na ⁇ ve-like CD4+ cells to na ⁇ ve-like CD8+ cells.
  • the first and/or second selections, or selections for sub-populations thereof, of the sample can be performed in a manner to result in an input cell composition with a desired ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ cells.
  • the ratio of CD4+ to CD8+ T cells in the sample is determined prior to performing the first and/or second selection from the sample. In certain instances, prior to performing the first and/or second selection, the ratio of na ⁇ ve-like CD4+ to na ⁇ ve-like CD8+ T cells in the sample is determined. Based on the particular ratio of the CD4+ to CD8+ T cells and/or na ⁇ ve-like CD4+ to CD8+ T cells in the sample, which can vary among samples, the particular mode of selection can be individualized to the sample, for example by sizing of chromatography columns or selection of amount or concentration of immunoaffinity reagents, to achieve the desired, fixed, or controlled ratio.
  • the relative level or frequency of various cell populations in a subject can be determined based on assessing surface expression of a marker or markers present on such populations or sub-populations.
  • a number of well-known methods for assessing expression level of surface markers or proteins may be used, such as detection by affinity-based methods, e.g., immunoaffinity-based methods, e.g., in the context of cell surface proteins, such as by flow cytometry.
  • the appropriate ratio for na ⁇ ve-like CD4+ and CD8+ T cells can vary depending on context, e.g., for example, for a particular disease, condition, or prior treatment of a subject from which cells are derived, and/or a particular antigen-specificity of the cells, relative representation among cells of a particular type (e.g., CD4+ cells) of various subpopulations, e.g., effector versus memory versus na ⁇ ve cells, and/or one or more conditions under which cells will be incubated, such as medium, stimulating agents, time of culture, buffers, oxygen content carbon dioxide content, antigen, cytokine, antibodies, and other components.
  • the ratio of na ⁇ ve-like CD4+ T cells and na ⁇ ve-like CD8+ T cells is determined based on known capacities of cell types in a normal or typical context, coupled with assessment of phenotypes or states of the cells or subject from which the cells are derived, and/or empirical evidence.
  • the separation and/or steps is carried out using immunomagnetic beads.
  • a cell sample containing CD4+ and CD8+ cells is contacted with magnetic beads containing a first immunoaffinity reagent that binds to CD4 or CD8 and magnetic beads containing a second immunoaffinity reagent that binds to the other of the CD4 or CD8.
  • the separation and/or steps can occur simultaneously and/or sequentially.
  • the first and/or second immunoaffinity reagent are present in the incubation composition at a sub-optimal yield concentration, whereby the enriched composition contains less than all, e.g., 70%, of the total CD4+ cells in the incubation composition and/or less than all, e.g., 70%, of the CD8+ cells in the incubation composition, thereby producing a composition enriched for CD4+ and CD8+ T cells.
  • the suboptimal yield concentration of the affinity reagent is a concentration below a concentration used or required to achieve an optimal or maximal yield of bound cells in a given selection or enrichment involving incubating cells with the reagent and recovering or separating cells having bound to the reagent ("yield,” for example, being the number of the cells so-recovered or selected compared to the total number of cells in the incubation that are targeted by the reagent or to which the reagent is specific or that have a marker for which the reagent is specific and capable of binding).
  • the suboptimal yield concentration generally is a concentration or amount of the reagent that in such process or step achieves less than all, e.g., no more than 70 % yield of bound cells, e.g., CD4+ and/or CD8+ T cells, upon recovery of the cells having bound to the reagent. In some instances, no more than at or about 50 %, 45 %, 40 %, 30 %, or 25 % yield is achieved by the suboptimal concentration of the affinity reagent.
  • the concentration may be expressed in terms of number or mass of particles or surfaces per cell and/or number of mass or molecules of agent (e.g., antibody, such as antibody fragment) per cell.
  • the suboptimal yield concentrations are sufficient to derive or achieve the fixed, controlled, and/or defined ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells.
  • one or more of such reagents is used at a concentration that is higher than one or more of the other such reagent(s), in order to bias the ratio of the cell type recognized by that reagent as compared to the cell type(s) recognized by the other(s).
  • the reagent specifically binding to the marker for which it is desired to bias the ratio may be included at a concentration (e.g., agent or mass per cells) that is increased by half, 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, or more, compared to other(s), depending on how much it is desired to increase the ratio.
  • concentration e.g., agent or mass per cells
  • the amount of immunoaffinity reagent is proportional to the approximate yield of enriched cells.
  • an appropriate amount or concentration of immunoaffinity reagents that depend on the desired ratio of the generated composition containing the enriched or selected CD4+ and CD8+ T cells can be determined as a matter of routine.
  • the separation and/or isolation steps are carried out using magnetic beads in which immunoaffinity reagents are reversibly bound, such as via a peptide ligand interaction with a streptavidin mutein as described in WO 2015/164675 .
  • Exemplary of such magnetic beads are Streptamers ® .
  • the separation and/or steps is carried out using magnetic beads, such as those commercially available from Miltenyi Biotec.
  • the first selection or enrichment of CD4+ and CD8+ cells from a sample are performed using immunoaffinity-based reagents that include at least a first and second affinity chromatography matrix, respectively, having immobilized thereon an antibody.
  • one or both of the first and/or second selection can employ a plurality of affinity chromatography matrices and/or antibodies, whereby the plurality of matrices and/or antibodies employed for the same selection, i.e. the first selection or the second selection, are serially connected.
  • the affinity chromatography matrix or matrices employed in a first and/or second selection adsorbs or is capable of selecting or enriching at least about 50 ⁇ 10 6 cells/mL, 100 ⁇ 10 6 cells/mL, 200 ⁇ 10 6 cells/mL or 400 ⁇ 10 6 cells/mL.
  • the adsorption capacity can be modulated based on the diameter and/or length of the column.
  • the culture-initiating ratio of the selected or enriched composition is achieved by choosing a sufficient amount of matrix and/or at a sufficient relative amount to achieve the culture-initiating ratio assuming based on, for example, the adsorption capacity of the column or columns for selecting cells.
  • the CD4+ T cells and CD8+ T cells have an equal or similar portion of na ⁇ ve-like cells, and the adsorption capacity of the matrix or matrices is the same between the first and second selection, e.g. is or is about 1 ⁇ 10 8 cells/mL for both, whereby enrichment or selection of cells in the first selection and second selection results in a composition containing a CD4+ cells to CD8+ cells with a na ⁇ ve-like CD4+ to CD8+ T cell ratio of or of about 1:1.
  • an appropriate volume, diameter or number of affinity matrix chromatography columns for the first and/or second selection depending on portions of na ⁇ ve-like cells and on the desired ratio of the generated input cell composition can be chosen or determined as a matter of routine.
  • the adsorption capacity of a column matrix or matrices is adjusted to account for differences in the frequency of a na ⁇ ve-like cells, e.g., naive like CD4+ or CD8+ cells, compared to the frequency of cells of the respective CD4+ or CD8+ parent population in the starting sample from the subject.
  • the relative level or frequency of various cell populations in a subject can be determined based on assessing surface expression of a marker or markers present on such populations or sub-populations.
  • a number of well-known methods for assessing expression level of surface markers or proteins may be used, such as detection by affinity-based methods, e.g., immunoaffinity-based methods, e.g., in the context of cell surface proteins, such as by flow cytometry.
  • affinity-based methods e.g., immunoaffinity-based methods, e.g., in the context of cell surface proteins, such as by flow cytometry.
  • na ⁇ ve-like cells e.g., na ⁇ ve-like CD4+ and/or CD8+ T cells
  • cell compositions e.g., CD4+ and/or CD8+ T cell compositions
  • sample e.g., a biological sample.
  • a na ⁇ ve-like T cell is a T cell that is positive for the expression of one or more markers that indicate that the cell is naive and/or is a na ⁇ ve-like cell.
  • a na ⁇ ve-like T cell is a cell that is positive for the expression of a marker that is associated with a naive or naive-like state in T cells.
  • a na ⁇ ve-like T cell is a T cell that is negative for the expression of one or more markers that indicates that the cell is not naive and/or is a not a na ⁇ ve-like cell.
  • a na ⁇ ve-like T cell is a cell that is negative for the expression of a marker that is associated with a non-naive or non-na ⁇ ve-like state in T cells.
  • a non-naive or non-na ⁇ ve-like state in a T cells includes, for example but not limited to, effector T (TEFF) cells, memory T cells, central memory T cells (TCM), effector memory T (TEM) cells, and combinations thereof.
  • TEFF effector T
  • TCM central memory T cells
  • TEM effector memory T
  • a na ⁇ ve-like T cell is positive for the expression of at least one, two, three, four, five, six, seven, eight, nine, ten, or more than ten markers that indicate that the cell is naive and/or is a na ⁇ ve-like cell, and/or is associated with a naive or na ⁇ ve-like state in T cells.
  • the markers are expressed on the cell surface.
  • the na ⁇ ve-like T cell is negative for the expression of at least one, two, three, four, five, six, seven, eight, nine, ten, or more than ten markers that indicate that the cell is non-na ⁇ ve and/or is a non-na ⁇ ve-like cell, and/or is associated with a non-naive or non-na ⁇ ve-like state in T cells.
  • Markers that indicate that the T cell is na ⁇ ve and/or is a na ⁇ ve-like T cell, and/or are associated with a naive or na ⁇ ve-like state in T cells include, but are not limited to, CD27, CD28, CD45RA, CD62L, and/or CCR7.
  • the na ⁇ ve-like T cell e.g., the na ⁇ ve-like CD4+ and/or CD8+ T cell
  • the na ⁇ ve-like T cell is positive for expression of CD27, CD28, CD45RA, and/or CCR7.
  • the na ⁇ ve-like T cell is positive for the surface expression of one or more of CD27, CD28, CD45RA, and/or CCR7.
  • the na ⁇ ve-like T cell e.g., the na ⁇ ve-like CD4+ and/or CD8+ T cell, is negative for expression of CD62L.
  • Markers that indicate that the cell is a non-naive and/or is a non-na ⁇ ve-like T cell, and/or are associated with a non-naive or non-na ⁇ ve-like state in T cells include, but are not limited to, CD25, CD45RO, CD56, KLRG1, and/or CD95.
  • the na ⁇ ve-like T cell e.g., a na ⁇ ve-like CD4+ and/or CD8+ T cell, is negative for expression of CD25, CD45RO, CD56, and/or KLRG1.
  • the na ⁇ ve-like T cell e.g., a na ⁇ ve-like CD4+ and/or CD8+ T cell
  • the na ⁇ ve-like T cell has low expression of CD95.
  • the na ⁇ ve-like T cell is negative for the surface expression of one or more of CD25, CD45RO, CD56, and/or KLRG1.
  • low expression of a marker associated with non-na ⁇ ve or non-na ⁇ ve-like cells is or includes at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% less expression than the expression of the marker in a cell that is a non-na ⁇ ve-like cells, and/or a cell that is positive for one or more markers that indicate that the cell is a non-naive and/or is a non-na ⁇ ve-like T cell, and/or are associated with a non-naive or non-naive-like state in T cells.
  • low expression of a marker associated with non-naive or non-na ⁇ ve-like cells is or includes at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% less expression than the expression of the marker in an effector T (T EFF ) cell, a memory T cell, a central memory T cell (T CM ), and/or an effector memory T (T EM ) cell.
  • T EFF effector T
  • T CM central memory T cell
  • T EM effector memory T
  • markers that indicate that the cell is a non-naive and/or is a non-na ⁇ ve-like T cell, and/or are associated with a non-naive or non-naive-like state in T cells include, one or more cytokines.
  • a non-naive or non-na ⁇ ve-like T cells is negative for the expression and/or the production of one or more of IL-2, IFN- ⁇ , IL-4, and IL-10.
  • the one or more cytokines are secreted.
  • the one or more cytokines are expressed internally by the non-na ⁇ ve-like T cells, for example, during or after treatment with an agent that prevents, inhibits, or reduces secretion.
  • a na ⁇ ve-like T cell is positive for the expression, e.g., surface expression, of CD45RA and CCR7.
  • a na ⁇ ve-like CD4+ T cell is positive for the expression, e.g., surface expression, of CD45RA and CCR7.
  • a na ⁇ ve-like CD8+ T cell is positive for the expression, e.g., surface expression, of CD45RA and CCR7.
  • a na ⁇ ve-like T cell is positive for the expression, e.g., surface expression, of CD45RA, CD27, and CCR7 and is negative for the expression, e.g., surface expression of CD45RO.
  • a na ⁇ ve-like CD4+ T cell is positive for the expression, e.g., surface expression, of CD45RA, CD27, and CCR7 and is negative for the expression, e.g., surface expression of CD45RO.
  • a na ⁇ ve-like CD8+ T cell is positive for the expression, e.g., surface expression, of CD45RA, CD27, and CCR7 and is negative for the expression, e.g., surface expression of CD45RO.
  • the CD4+ and/or CD8+ T cells are viable cells. In certain instances, the CD4+ and/or CD8+ T cells are viable na ⁇ ve-like cells. The viable cell is positive for the expression of a marker that indicates that the cell undergoes normal functional cellular processes and/or has not undergone or is not under the process of undergoing necrosis or programmed cell death. In some instances, viability can be assessed by the redox potential of the cell, the integrity of the cell membrane, or the activity or function of mitochondria. In some instances, viability is the absence of a specific molecule associated with cell death, or the absence of the indication of cell death in an assay.
  • cell viability is assessed with an assay that may include, but is not limited to, dye uptake assays (e.g., calcein AM assays), XTT cell viability assays, and dye exclusion assays (e.g., trypan blue, Eosin, or propidium dye exclusion assays).
  • a viable cell has negative expression of one or more apoptotic markers, e.g., annexin V or active Caspase 3.
  • the viable cell is negative for the expression or activation of one or more apoptosis marker that may include, but are not limited to, a caspase, e.g., caspase 2, caspase 3, caspase 6, caspase 7, caspase 8, caspase 9, and caspase 10, Bcl-2 family members, e.g., Bax, Bad, and Bid, Annexin V, and/or TUNEL staining.
  • a caspase e.g., caspase 2, caspase 3, caspase 6, caspase 7, caspase 8, caspase 9, and caspase 10, Bcl-2 family members, e.g., Bax, Bad, and Bid, Annexin V, and/or TUNEL staining.
  • expression is or includes an amount, level, concentration, and/or presence of the marker.
  • the marker is polypeptide.
  • the marker is an mRNA.
  • the expression is or includes an amount, level, concentration, and/or presence of a polypeptide, e.g., the marker polypeptide.
  • expression is or includes an amount, level, concentration, and/or presence of the marker on or exposed on the cell surface or within the cell membrane.
  • expression is or includes an amount, level, concentration, and/or presence of the marker on or exposed on the cell surface or within the cell membrane.
  • the expression is or includes internal expression, e.g., an amount, level, concentration, and/or presence of the marker within the cell internally, such as within the cytosol, nucleus, endoplasmic reticulum, and/or the Golgi apparatus.
  • the markers are measured, assessed, and/or quantified by performing an in vitro assay.
  • the in vitro assay is an immunoassay, an aptamer-based assay, a histological or cytological assay, or an mRNA expression level assay.
  • the in vitro assay used can be an enzyme-linked immunosorbent assay (ELISA), immunoblotting, immunoprecipitation, radioimmunoassay (RIA), immunostaining, flow cytometry assay, surface plasmon resonance (SPR), chemiluminescence assay, lateral flow immunoassay, inhibition assay or avidity assay.
  • the expression of the markers is measured, assessed, and/or quantified by RNA-seq. In particular instances, the expression of the markers is measured, assessed, and/or quantified by immunostaining techniques. In particular instances, the expression of the markers is measured, assessed, and/or quantified by flow cytometry analysis. In some instances, the expression of the markers is measured, assessed, and/or quantified by internal cytokine staining.
  • the markers are measured, assessed, and/or quantified in cells of the CD4+ T cell composition.
  • at least 1%, at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 95%,at least 97%, or at least 99% of the CD4+ T cells are na ⁇ ve-like CD4+ T cells.
  • the CD4+ T cells are na ⁇ ve-like CD4+ T cells.
  • the na ⁇ ve-like CD4+ T cells are viable na ⁇ ve-like CD4+ T cells.
  • the markers are measured, assessed, and/or quantified in cells of the CD8+ T cell composition.
  • at least 1%, at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 95%,at least 97%, or at least 99% of the CD8+ T cells are na ⁇ ve-like CD8+ T cells.
  • the CD8+ T cells are na ⁇ ve-like CD4+ T cells.
  • the na ⁇ ve-like CD8+ T cells are viable na ⁇ ve-like CD8+ T cells.
  • cells from a composition of CD4+ T cells are mixed or combined with cells from a composition of CD8+ T cells in amounts and/or proportions sufficient to produce an input cell composition with a ratio of CD4+ na ⁇ ve-like T cells to CD8+ na ⁇ ve-like T cells between 10:1 and 0.05:1, between 8:1 and 0.1:1, between 5:1 and 0.2:1, between 2.5:1 and 0.25:1, between 2.2:1 and 0.8:1, between 2:1 and 0.5:1, or between 1.5:1 and 1:1, inclusive.
  • the cells are mixed in amounts and/or proportions sufficient to a ratio of CD4+ na ⁇ ve-like T cells to CD8+ na ⁇ ve-like T cells of between 2.2:1 and 0.8:1, inclusive. In certain instances, the cells are mixed or combined to a ratio of CD4+ na ⁇ ve-like T cells to CD8+ na ⁇ ve-like T cells of or of about 2.2:1, 2.1:1, 2.0:1, 1.9:1, 1.8:1, 1.7:1, 1.6:1, 1.5:1, 1.4:1, 1.3:1, 1.2:1, 1.1:1, 1.0:1, 0.9:1, or 0.8:1. In certain instances, the cells are mixed or combined to a ratio of or of about 1.1:1.
  • between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total or total viable CD4+ T cells are mixed or combined with an amount of or about of between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total or total viable CD8+ T cells to produce an input cell composition with a defined ratio of CD4+ na ⁇ ve-like T cells to CD8+ na ⁇ ve-like T cells.
  • between 5 ⁇ 10 5 and 1 ⁇ 10 10 , 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 na ⁇ ve-like CD4+ T cells are mixed or combined with an amount of or about of between 5 ⁇ 10 5 and 1 ⁇ 10 10 , 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 na ⁇ ve-like CD8+ T cells to produce an input cell composition with a defined ratio of CD4+ na ⁇ ve-like T cells to CD8+ na ⁇ ve-like T cells.
  • the ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells of the input cell composition has been adjusted, changed, and/or altered compared to the ratio of the na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells of a sample, e.g., a biological sample.
  • the ratio of na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells is or is about or is at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 100%, 1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 4-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold adjusted, changed, or altered from the biological sample.
  • the sample is the sample from where cells of the input cell composition were derived, isolated, selected, and/or obtained.
  • the input cell composition contains a ratio, e.g., a defined, controlled, and/or fixed ratio, of CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells.
  • the ratio of CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells is between 10:1 to 0.05:1, between 8:1 and 0.1:1, between 5:1 and 0.2:1, between 2.5:1 and 0.25:1, between 2.2:1 and 0.8:1, between 2:1 and 0.5:1, or between 1.5:1 and 1:1, inclusive.
  • the ratio of CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells is between 2:1 and 0.8:1, between 1.6:1 and 0.8:1, between 1.4:1 and 0.8:1, between 1.2:1 and 0.8:for between 1.2:1 and 0.8:1, inclusive. In some instances, the ratio is between 2.2:1 and 0.8:1, inclusive.
  • the ratio of CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells is or is about 2.2:1, 2.1:1, 2.0:1, 1.9:1, 1.8:1, 1.7:1, 1.6:1, 1.5:1, 1.4:1, 1.3:1, 1.2:1, 1.1:1, 1.0:1, 0.9:1, or 0.8:1. In certain instances, the ratio is or is about 1.1:1.
  • the input cell composition has an amount of or about of 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1.0 ⁇ 10 8 , 1.1 ⁇ 10 8 , 1.2 ⁇ 10 8 , 1.3 ⁇ 10 8 , 1.4 ⁇ 10 8 , 1.5 ⁇ 10 8 , 1.6 ⁇ 10 8 , 1.7 ⁇ 10 8 , 1.8 ⁇ 10 8 , 1.9 ⁇ 10 8 , 2.0 ⁇ 10 8 , 2.1 ⁇ 10 8 , 2.2 ⁇ 10 8 , 2.3 ⁇ 10 8 , 2.4 ⁇ 10 8 , 2.5 ⁇ 10 8 , 2.6 ⁇ 10 8 , 2.7 ⁇ 10 8 , 2.8 ⁇ 10 8 , 2.9 ⁇ 10 8 , 3.0 ⁇ 10 8 , 3.5 ⁇ 10 8 , 4.0 ⁇ 10 8 , 4.5 ⁇ 10 8 , 5 ⁇ 10 8 , 5 ⁇ 10 8 , or 1 ⁇ 10 9 total cells or
  • the input cell composition has an amount of or about of 5 ⁇ 10 5 , 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1.0 ⁇ 10 8 , 1.1 ⁇ 10 8 , 1.2 ⁇ 10 8 , 1.3 ⁇ 10 8 , 1.4 ⁇ 10 8 , 1.5 ⁇ 10 8 , 1.6 ⁇ 10 8 , 1.7 ⁇ 10 8 , 1.8 ⁇ 10 8 , 1.9 ⁇ 10 8 , 2.0 ⁇ 10 8 , 2.1 ⁇ 10 8 , 2.2 ⁇ 10 8 , 2.3 ⁇ 10 8 , 2.4 ⁇ 10 8 , 2.5 ⁇ 10 8 , 2.6 ⁇ 10 8 , 2.7 ⁇ 10 8 , 2.8 ⁇ 10 8 , 2.9 ⁇ 10 8 , 3.0 ⁇ 10 8 , 3.5 ⁇ 10 8 , 4.0 ⁇ 10 8 , 4.5 ⁇ 10 8 , 5 ⁇ 10 8 , 5 ⁇ 10 8 , or 1
  • the input cell composition has an amount of or about of 5 ⁇ 10 5 , 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1.0 ⁇ 10 8 , 1.1 ⁇ 10 8 , 1.2 ⁇ 10 8 , 1.3 ⁇ 10 8 , 1.4 ⁇ 10 8 , 1.5 ⁇ 10 8 , 1.6 ⁇ 10 8 , 1.7 ⁇ 10 8 , 1.8 ⁇ 10 8 , 1.9 ⁇ 10 8 , 2.0 ⁇ 10 8 , 2.1 ⁇ 10 8 , 2.2 ⁇ 10 8 , 2.3 ⁇ 10 8 , 2.4 ⁇ 10 8 , 2.5 ⁇ 10 8 , 2.6 ⁇ 10 8 , 2.7 ⁇ 10 8 , 2.8 ⁇ 10 8 , 2.9 ⁇ 10 8 , 3.0 ⁇ 10 8 , 3.5 ⁇ 10 8 , 4.0 ⁇ 10 8 , 4.5 ⁇ 10 8 , 5 ⁇ 10 8 , 5 ⁇ 10 8 , or 1
  • the input cell composition has between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total cells or total viable cells.
  • the input cell composition has an amount of or about of between 5 ⁇ 10 5 and 1 ⁇ 10 10 , 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 cells that express CD4 or CD8.
  • the input cell composition has an amount of or about of between 5 ⁇ 10 5 and 1 ⁇ 10 10 , 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 CD45RA+/CCR7+/CD4+ and CD45RA+/CCR7+/CD8+ T cells.
  • the input cell composition has or contains at least 1%, at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70% , at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or 100% or about 100% CD45RA+/CCR7+ cells.
  • the input cell composition contains or includes no more than 100%, no more than 99%, no more than 98%, no more than 97%, no more than 96%, no more than 95%, no more than 90%, or no more than 85% CD45RA+/CCR7+ cells.
  • between 5 ⁇ 10 5 and 1 ⁇ 10 10 , 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total or total viable CD4+ T cells are mixed or combined with an amount of or about of between 5 ⁇ 10 5 and 1 ⁇ 10 10 , 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total or total viable CD8+ T cells to produce an input cell composition with a defined ratio of CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells.
  • the ratio of CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells of the input cell composition has been adjusted, changed, and/or altered compared to the ratio of the CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells of a sample, e.g., a biological sample.
  • the ratio of CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells is or is about or is at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 100%, 1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 4-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold adjusted, changed, or altered from the biological sample.
  • the sample is the sample from where cells of the input cell composition were derived, isolated, selected, and/or obtained.
  • the input cell composition contains a ratio, e.g., a defined, controlled, and/or fixed ratio, of CD27+/CCR7+/CD4+ T cells to CD27+/CCR7+/CD8+ T cells.
  • the ratio of CD27+/CCR7+/CD4+ T cells to CD27+/CCR7+/CD8+ T cells is between 10:1 and 0.05:1, between 8:1 and 0.1:1, between 5:1 and 0.2:1, between 2.5:1 and 0.25:1, between 2.2:1 and 0.8:1, between 2:1 and 0.5:1, or between 2:1 and 1:1, inclusive.
  • the ratio of CD27+/CCR7+/CD4+ T cells to CD27+/CCR7+/CD8+ T cells is between 2:1 and 0.8:1, between 1.8:1 and 1:1, between 1.8:1 and 1.2:1, between 1.2:1 and 1.4:1,or between 1.8:1 and 1.6:1, inclusive. In some instances, the ratio is between 1.8:1 and 1.6:1, inclusive. In certain instances, the ratio of CD27+/CCR7+/CD4+ T cells to CD27+/CCR7+/CD8+ T cells is or is about 2.2:1, 2.1:1, 2.0:1, 1.9:1, 1.8:1, 1.7:1, 1.69:1, 1.6:1, 1.5:1, 1.4:1, or 1.3:1. In certain instances, the ratio is or is about 1.69:1.
  • the input cell composition has an amount of or about of 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1.0 ⁇ 10 8 , 1.1 ⁇ 10 8 , 1.2 ⁇ 10 8 , 1.3 ⁇ 10 8 , 1.4 ⁇ 10 8 , 1.5 ⁇ 10 8 , 1.6 ⁇ 10 8 , 1.7 ⁇ 10 8 , 1.8 ⁇ 10 8 , 1.9 ⁇ 10 8 , 2.0 ⁇ 10 8 , 2.1 ⁇ 10 8 , 2.2 ⁇ 10 8 , 2.3 ⁇ 10 8 , 2.4 ⁇ 10 8 , 2.5 ⁇ 10 8 , 2.6 ⁇ 10 8 , 2.7 ⁇ 10 8 , 2.8 ⁇ 10 8 , 2.9 ⁇ 10 8 , 3.0 ⁇ 10 8 , 3.5 ⁇ 10 8 , 4.0 ⁇ 10 8 , 4.5 ⁇ 10 8 , 5 ⁇ 10 8 , 5 ⁇ 10 8 , or 1 ⁇ 10 9 total cells or
  • the input cell composition has an amount of or about of 5 ⁇ 10 5 , 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1.0 ⁇ 10 8 , 1.1 ⁇ 10 8 , 1.2 ⁇ 10 8 , 1.3 ⁇ 10 8 , 1.4 ⁇ 10 8 , 1.5 ⁇ 10 8 , 1.6 ⁇ 10 8 , 1.7 ⁇ 10 8 , 1.8 ⁇ 10 8 , 1.9 ⁇ 10 8 , 2.0 ⁇ 10 8 , 2.1 ⁇ 10 8 , 2.2 ⁇ 10 8 , 2.3 ⁇ 10 8 , 2.4 ⁇ 10 8 , 2.5 ⁇ 10 8 , 2.6 ⁇ 10 8 , 2.7 ⁇ 10 8 , 2.8 ⁇ 10 8 , 2.9 ⁇ 10 8 , 3.0 ⁇ 10 8 , 3.5 ⁇ 10 8 , 4.0 ⁇ 10 8 , 4.5 ⁇ 10 8 , 5 ⁇ 10 8 , 5 ⁇ 10 8 , or 1
  • the input cell composition has an amount of or about of 5 ⁇ 10 5 , 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1.0 ⁇ 10 8 , 1.1 ⁇ 10 8 , 1.2 ⁇ 10 8 , 1.3 ⁇ 10 8 , 1.4 ⁇ 10 8 , 1.5 ⁇ 10 8 , 1.6 ⁇ 10 8 , 1.7 ⁇ 10 8 , 1.8 ⁇ 10 8 , 1.9 ⁇ 10 8 , 2.0 ⁇ 10 8 , 2.1 ⁇ 10 8 , 2.2 ⁇ 10 8 , 2.3 ⁇ 10 8 , 2.4 ⁇ 10 8 , 2.5 ⁇ 10 8 , 2.6 ⁇ 10 8 , 2.7 ⁇ 10 8 , 2.8 ⁇ 10 8 , 2.9 ⁇ 10 8 , 3.0 ⁇ 10 8 , 3.5 ⁇ 10 8 , 4.0 ⁇ 10 8 , 4.5 ⁇ 10 8 , 5 ⁇ 10 8 , 5 ⁇ 10 8 , or 1
  • the input cell composition has between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total cells or total viable cells.
  • the input cell composition has an amount of or about of between 5 ⁇ 10 5 and 1 ⁇ 10 10 , 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 cells that express CD4 or CD8.
  • the input cell composition has an amount of or about of between 5 ⁇ 10 5 and 1 ⁇ 10 10 , 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 CD27+/CCR7+/CD4+ and CD27+/CCR7+/CD8+ T cells.
  • the input cell composition has or contains at least 1%, at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70% , at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or 100% or about 100% CD27+/CCR7+ cells.
  • the input cell composition contains or includes no more than 100%, no more than 99%, no more than 98%, no more than 97%, no more than 96%, no more than 95%, no more than 90%, or no more than 85% CD27+/CCR7+ cells.
  • between 5 ⁇ 10 5 and 1 ⁇ 10 10 , 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total or total viable CD4+ T cells are mixed or combined with an amount of or about of between 5 ⁇ 10 5 and 1 ⁇ 10 10 , 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total or total viable CD8+ T cells to produce an input cell composition with a defined ratio of CD27+/CCR7+/CD4+ T cells to CD27+/CCR7+/CD8+ T cells.
  • the ratio of CD27+/CCR7+/CD4+ T cells to CD27+/CCR7+/CD8+ T cells of the input cell composition has been adjusted, changed, and/or altered compared to the ratio of the CD27+/CCR7+/CD4+ T cells to CD27+/CCR7+/CD8+ T cells of a sample, e.g., a biological sample.
  • the ratio of CD27+/CCR7+/CD4+ T cells to CD27+/CCR7+/CD8+ T cells is or is about or is at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 100%, 1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 4-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold adjusted, changed, or altered from the biological sample.
  • the sample is the sample from where cells of the input cell composition were derived, isolated, selected, and/or obtained.
  • the input cell composition contains a ratio, e.g., a defined, controlled, and/or fixed ratio, of CD62L-/CCR7+/CD4+ T cells to CD62L-/CCR7+/CD8+ T cells.
  • the ratio of CD62L-/CCR7+/CD4+ T cells to CD62L-/CCR7+/CD8+ T cells is between 10:1 and 0.05:1, between 8:1 and 0.1:1, between 5:1 and 0.2:1, between 2.5:1 and 0.25:1, between 2.2:1 and 0.8:1, between 2:1 and 0.5:1, or between 1.5:1 and 1:1, inclusive.
  • the ratio of CD62L-/CCR7+/CD4+ T cells to CD62L-/CCR7+/CD8+ T cells is between 2:1 and 0.8:1, between 1.6:1 and 0.8:1, between 1.4:1 and 0.8:1, between 1.2:1 and 0.8:1,or between 1.2:1 and 0.8:1, inclusive. In some instances, the ratio is between 2.2:1 and 0.8:1, inclusive.
  • the ratio of CD62L-/CCR7+/CD4+ T cells to CD62L-/CCR7+/CD8+ T cells is or is about 2.2:1, 2.1:1, 2.0:1, 1.9:1, 1.8:1, 1.7:1, 1.6:1, 1.5:1, 1.4:1, 1.3:1, 1.2:1, 1.1:1, 1.0:1, 0.9:1, or 0.8:1. In certain instances, the ratio is or is about 1.1:1.
  • the input cell composition has between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total cells or total viable cells. In certain instances, the input cell composition has an amount of or about of between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 cells that express CD4 or CD8.
  • the input cell composition has an amount of or about of between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 CD62L-/CCR7+/CD4+ and CD62L-/CCR7+/CD8+ T cells.
  • the input cell composition has or contains at least 1%, at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70% , at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or 100% or about 100% CD62L-/CCR7+ cells.
  • the input cell composition contains or includes no more than 100%, no more than 99%, no more than 98%, no more than 97%, no more than 96%, no more than 95%, no more than 90%, or no more than 85% CD62L-/CCR7+ cells.
  • between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total or total viable CD4+ T cells are mixed or combined with an amount of or about of between 1 ⁇ 10 6 and 1 ⁇ 10 10 , between 1 ⁇ 10 7 and 1 ⁇ 10 9 , between 5 ⁇ 10 7 and 5 ⁇ 10 8 , or between 1 ⁇ 10 8 and 3 ⁇ 10 8 total or total viable CD8+ T cells to produce an input cell composition with a defined ratio of CD62L-/CCR7+/CD4+ T cells to CD62L-/CCR7+/CD8+ T cells.
  • the ratio of CD62L-/CCR7+/CD4+ T cells to CD62L-/CCR7+/CD8+ T cells of the input cell composition has been adjusted, changed, and/or altered compared to the ratio of the CD62L-/CCR7+/CD4+ T cells to CD62L-/CCR7+/CD8+ T cells of a sample, e.g., a biological sample.
  • the ratio of CD62L-/CCR7+/CD4+ T cells to CD62L-/CCR7+/CD8+ T cells is or is about or is at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 100%, 1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 4-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold adjusted, changed, or altered from the biological sample.
  • the sample is the sample from where cells of the input cell composition were derived, isolated, selected, and/or obtained.
  • producing, generating, and/or making an input cell composition includes one or more steps of mixing or combining cells of a CD4+ T cell composition with cells of a CD8+ T cell compositions to produce an input cell composition with a ratio of between 2.2:1 to 0.8:1 CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells.
  • number, number per volume, number per weight, and/or the amount, level, or percentage of CD45RA+/CCR7+ cells are measured, assessed, and/or quantified in the CD4+ T cell composition and the CD8+ T cell composition prior to the mixing or combining.
  • the amount, level, number, number per volume, number per weight, and/or percentage of CD45RA+/CCR7+ cells are measured, assessed, and/or quantified by detecting CD45RA+;CCR7+ T cells.
  • the input cell composition has a ratio of between 2.2:1 to 0.8:1 CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells.
  • the input cell composition has a ratio of or of about 1.1:1 CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells.
  • producing, generating, and/or making an input cell composition includes one or more steps of mixing or combining cells of a CD4+ T cell composition with cells of a CD8+ T cell compositions to produce an input cell composition with a ratio of between 2.4:1 to 1:1 CD27+/CCR7+/CD4+ T cells to CD27+/CCR7+/CD8+ T cells.
  • number, number per volume, number per weight, and/or the amount, level, or percentage of CD27+/CCR7+ cells are measured, assessed, and/or quantified in the CD4+ T cell composition and the CD8+ T cell composition prior to the mixing or combining.
  • the amount, level, number, number per volume, number per weight, and/or percentage of CD27+/CCR7+ cells are measured, assessed, and/or quantified by detecting CD45RA+;CCR7+ T cells.
  • the input cell composition has a ratio of between 2.4:1 to 1:1 CD27+/CCR7+/CD4+ T cells to CD27+/CCR7+/CD8+ T cells.
  • the input cell composition has a ratio of or of about 1.69:1 CD27+/CCR7+/CD4+ T cells to CD27+/CCR7+/CD8+ T cells.
  • producing, generating, and/or making an input cell composition includes one or more steps of mixing or combining cells of a CD4+ T cell composition with cells of a CD8+ T cell compositions to produce an input cell composition with a ratio of between 2.2:1 to 0.8:1 CD62L-/CCR7+/CD4+ T cells to CD62L-/CCR7+/CD8+ T cells.
  • number, number per volume, number per weight, and/or the amount, level, or percentage of CD62L-/CCR7+ cells are measured, assessed, and/or quantified in the CD4+ T cell composition and the CD8+ T cell composition prior to the mixing or combining.
  • the amount, level, number, number per volume, number per weight, and/or percentage of CD62L-/CCR7+ cells are measured, assessed, and/or quantified by detecting CD62L-/CCR7+ T cells.
  • the input cell composition has a ratio of between 2.2:1 to 0.8:1 CD62L-/CCR7+/CD4+ T cells to CD62L-/CCR7+/CD8+ T cells. In some instances, the input cell composition has a ratio of or of about 1.1:1 CD62L-/CCR7/CD4+ T cells to CD62L-/CCR7/CD8+ T cells.
  • producing, generating, and/or making an input cell composition includes one or more steps of mixing or combining cells of a CD4+ T cell composition with cells of a CD8+ T cell compositions to produce an input cell composition with a ratio of between 2.2:1 to 0.8:1 na ⁇ ve-like CD4+ T cells to na ⁇ ve-like CD8+ T cells.
  • number, number per volume, number per weight, and/or the amount, level, or percentage of na ⁇ ve-like cells are measured, assessed, and/or quantified in the CD4+ T cell composition and the CD8+ T cell composition prior to the mixing or combining.
  • the amount, level, number, number per volume, number per weight, and/or percentage of na ⁇ ve-like cells are measured, assessed, and/or quantified by detecting CD45RA+;CCR7+ T cells.
  • the input cell composition has a ratio of between 2.2:1 to 0.8:1 CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells.
  • the input cell composition has a ratio of or of about 1.1:1 CD45RA+/CCR7+/CD4+ T cells to CD45RA+/CCR7+/CD8+ T cells.
  • the cells are incubated and/or cultured prior to, in connection with, and/or subsequent to a process for genetic engineering.
  • the cells are incubated and/or cultured prior to, during, or immediately after one or more steps associated with the genetic engineering protocol or process.
  • the incubation steps can include culture, cultivation, stimulation, activation, and/or propagation.
  • the incubation and/or engineering may be carried out in a culture vessel, such as a unit, chamber, well, column, tube, tubing set, valve, vial, culture dish, bag, or other container for culture or cultivating cells.
  • the compositions or cells are incubated in the presence of stimulating conditions or a stimulatory agent. Such conditions include those designed to induce proliferation, expansion, activation, and/or survival of cells in the population, to mimic antigen exposure, and/or to prime the cells for genetic engineering, such as for the introduction of a recombinant antigen receptor.
  • methods disclosed herein include on or more steps of incubating cells under stimulating or activating conditions.
  • the cells are incubated for or for about 15 minutes, 30 minutes, 45 minutes, 60 minutes, 90 minutes, 120 minutes, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 12 hours, 16 hours, 18 hours, 24 hours, 36 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or more than 7 days.
  • the cells are incubated for between 5 minutes and 90 minutes, between 1 hour and 4 hours, between 2 hours and 8 hours, between 6 hours and 24 hours, between 12 hours and 48 hours, between 16 hours and 32 hours, between 18 hours and 30 hours, between 1 day and 4 days, or between 2 days and 7 days.
  • the cells are incubated for between 2 and 15 days, between 2 and 12 days, between 2 and 10 days, between 2 and 8 days, between 2 and 6 days, between 2 and 4 days, between 4 and 12 days, between 4 and 10 days, between 4 and 8 days, between 4 and 6 days, between 6 and 12 days, between 6 and 10 days, between 6 and 8 days, between 8 and 12 days, between 8 and 10 days, or between 10 and 12 days.
  • the cells are incubated for at least 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, or more than 14 days.
  • the cells are genetically engineered by a method or process that includes one or more steps for gene transfer.
  • the gene transfer is accomplished by first stimulating the cells, such as by incubating the cells under stimulating conditions, e.g., incubating the cells with a stimulus that induces a response such as proliferation, survival, and/or activation, followed by the gene transfer.
  • the cells are incubated under stimulating conditions for the purposes of activating the cells prior to the gene transfer.
  • the gene transfer is or includes transduction of the activated cells.
  • the stimulating agents or conditions induce and/or are capable of inducing primary signal, signaling, stimulation, activation and/or expansion of the cells.
  • the cells e.g., the cells of the input cell composition are incubated under stimulating or activating conditions prior to gene transfer. In some instances, the cells are incubated under stimulating or activating conditions for or for about 15 minutes, 30 minutes, 45 minutes, 60 minutes, 90 minutes, 120 minutes, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 12 hours, 16 hours, 18 hours, 24 hours, 36 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or more than 7 days.
  • the cells of the input cell composition are incubated under stimulating or activating conditions for between 5 minutes and 90 minutes, between 1 hour and 4 hours, between 2 hours and 8 hours, between 6 hours and 24 hours, between 12 hours and 48 hours, between 16 hours and 32 hours, between 18 hours and 30 hours, between 1 day and 4 days, or between 2 days and 7 days prior to gene transfer.
  • the cells of the input cell composition are incubated under stimulating or activating conditions for between 2 and 15 days, between 2 and 12 days, between 2 and 12 days, between 2 and 8 days, between 2 and 6 days, between 2 and 4 days, between 4 and 12 days, between 4 and 10 days, between 4 and 8 days, between 4 and 6 days, between 6 and 12 days, between 6 and 10 days, between 6 and 8 days, between 8 and 12 days, between 8 and 10 days, or between 10 and 12 days.
  • the cells are incubated for at least 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, or more than 14 days.
  • the cells are incubated under stimulating or activating conditions during some or all of a gene transfer.
  • the cells are incubated under stimulating conditions during all or a portion of transducing or transfecting the cells to introduce a heterologous nucleic acid.
  • the cells are incubated under stimulating or activating conditions in the presence of a viral vector, e.g., a retroviral vector such a gammaretroviral vector or a lentiviral vector.
  • the cells are incubated under stimulating or activating conditions after, e.g., immediately after the gene transfer.
  • the cells are incubated under stimulating or activating conditions after the gene transfer to expand the cells e.g., to expand the cells into numbers that are sufficient for clinical applications.
  • the cells are incubated after the gene transfer under stimulating or activating conditions for or for about 15 minutes, 30 minutes, 45 minutes, 60 minutes, 90 minutes, 120 minutes, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 12 hours, 16 hours, 18 hours, 24 hours, 36 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or more than 7 days.
  • the cells composition are incubated after the gene transfer under stimulating or activating conditions for between 5 minutes and 90 minutes, between 1 hour and 4 hours, between 2 hours and 8 hours, between 6 hours and 24 hours, between 12 hours and 48 hours, between 16 hours and 32 hours, between 18 hours and 30 hours, between 1 day and 4 days, or between 2 days and 7 days prior to gene transfer.
  • the cells of the composition are incubated after the gene transfer under stimulating or activating conditions for between 2 and 15 days, between 2 and 12 days, between 2 and 12 days, between 2 and 8 days, between 2 and 6 days, between 2 and 4 days, between 4 and 12 days, between 4 and 10 days, between 4 and 8 days, between 4 and 6 days, between 6 and 12 days, between 6 and 10 days, between 6 and 8 days, between 8 and 12 days, between 8 and 10 days, or between 10 and 12 days.
  • the cells are incubated for at least 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, or more than 14 days.
  • the cells are expanded for an amount of time sufficient to generate a suitable number of cells for a clinical application.
  • the conditions can include one or more of particular media, temperature, oxygen content, carbon dioxide content, time, agents, e.g., nutrients, amino acids, antibiotics, ions, and/or stimulatory factors, such as cytokines, chemokines, antigens, binding partners, fusion proteins, recombinant soluble receptors, and any other agents designed to activate the cells.
  • agents e.g., nutrients, amino acids, antibiotics, ions, and/or stimulatory factors, such as cytokines, chemokines, antigens, binding partners, fusion proteins, recombinant soluble receptors, and any other agents designed to activate the cells.
  • stimulatory conditions or agents induce and/or are capable of inducing primary signal, signaling, stimulation, activation and/or expansion of the cells.
  • the stimulating conditions or agents include one or more agent, e.g., ligand, which is capable of activating an intracellular signaling domain of a TCR complex.
  • the agent turns on or initiates TCR/CD3 intracellular signaling cascade in a T cell.
  • agents can include antibodies, such as those specific for a TCR, e.g. anti-CD3.
  • the stimulating conditions include one or more agent, e.g. ligand, which is capable of stimulating a costimulatory receptor, e.g., anti-CD28.
  • agents and/or ligands may be, bound to solid support such as a bead, and/or one or more cytokines.
  • the expansion method may further comprise the step of adding anti-CD3 and/or anti-CD28 antibody to the culture medium (e.g., at a concentration of at least about 0.5 ng/ml).
  • the stimulating agents include IL-2, IL-15 and/or IL-7.
  • the agent is an antibody that specifically binds to the recombinant receptor.
  • the recombinant receptor is an antigen binding receptor, e.g., a CAR, and the agent is an anti-idiotype antibody that specifically binds to the antigen binding receptor.
  • the agents and/or ligands are attached to magnetic beads, such as paramagnetic beads (e.g., such as Dynalbeads or MACS beads).
  • the beads may be removed from the cells by placing the cells in a magnetic field, thus removing and/or separating the beads from the cells, e.g., debeading the cells.
  • the beads are removed immediately after an incubation to activate the cells.
  • the beads are removed prior to gene delivery, for example by transduction or transfection.
  • the beads are removed during or immediately after the gene delivery.
  • the beads are removed prior to or during an incubation to expanding the cells.
  • the beads are removed prior to harvesting and/or cryoprotecting the cells.
  • incubation is carried out in accordance with techniques such as those described in US Patent No. 6,040,177 to Riddell et al. , Klebanoff et al. (2012) J Immunother. 35(9): 651-660 , Terakura et al. (2012) Blood.1:72-82 , and/or Wang et al. (2012) J Immunother. 35(9):689-701 .
  • the T cells are expanded by adding to a culture-initiating composition feeder cells, such as non-dividing peripheral blood mononuclear cells (PBMC), (e.g., such that the resulting population of cells contains at least about 5, 10, 20, or 40 or more PBMC feeder cells for each T lymphocyte in the initial population to be expanded); and incubating the culture (e.g. for a time sufficient to expand the numbers of T cells).
  • PBMC peripheral blood mononuclear cells
  • the non-dividing feeder cells can comprise gamma-irradiated PBMC feeder cells.
  • the PBMC are irradiated with gamma rays in the range of about 3000 to 3600 rads to prevent cell division.
  • the feeder cells are added to culture medium prior to the addition of the populations of T cells.
  • the stimulating conditions include temperature suitable for the growth of human T lymphocytes, for example, at least about 25 degrees Celsius, generally at least about 30 degrees, and generally at or about 37 degrees Celsius.
  • the incubation may further comprise adding non-dividing EBV-transformed lymphoblastoid cells (LCL) as feeder cells.
  • LCL can be irradiated with gamma rays in the range of about 6000 to 10,000 rads.
  • the LCL feeder cells in some aspects is provided in any suitable amount, such as a ratio of LCL feeder cells to initial T lymphocytes of at least about 10:1.
  • antigen-specific T cells such as antigen-specific CD4+ and/or CD8+ T cells
  • antigen-specific T cell lines or clones can be generated to cytomegalovirus antigens by isolating T cells from infected subjects and stimulating the cells in vitro with the same antigen.
  • the disclosed methods involve generating or producing a composition of engineered cells, e.g., a cell composition containing cells that express a recombinant antigen receptor.
  • a composition of engineered cells e.g., a cell composition containing cells that express a recombinant antigen receptor.
  • recombinant receptors e.g., CARs or TCRs
  • Exemplary methods include those for transfer of nucleic acids encoding the receptors, including via viral, e.g., retroviral or lentiviral, transduction, transposons, and electroporation.
  • cells of the input cell composition are contacted with and/or introduced with one or more agents containing a nucleic acid, for example that encode a recombinant receptor such as a CAR.
  • the agent is a vector, e.g., a viral vector, a plasmid, or a transposon.
  • the cells are CD4+ T cells.
  • the cells are CD8+ T cells.
  • the introducing and/or contacting the cells with the nucleic acid results in engineered cells with a defined, desired, or fixed ratio of engineered CD4+ T cells to engineered CD8+ T cells.
  • the nucleic acids are heterologous, i.e., normally not present in a cell or sample obtained from the cell, such as one obtained from another organism or cell, which for example, is not ordinarily found in the cell being engineered and/or an organism from which such cell is derived.
  • the nucleic acids are not naturally occurring, such as a nucleic acid not found in nature, including one comprising chimeric combinations of nucleic acids encoding various domains from multiple different cell types.
  • the preparation methods include steps for freezing, e.g., cryopreserving, the cells, either before, such as immediately before, during, or after, such as immediately after, any of the steps or stages for genetic engineering and/or producing an output cell composition that contains genetically engineered cells.
  • the cells are cryofrozen before, during, or after the steps of isolating or selecting the cells, mixing or combining cells into an input cell composition, incubation, activation, gene transfer, transduction, transfection, expansion, and/or harvesting. In some instances, all or a portion of the cells are collected for freezing.
  • a portion of the cells are collecting before, during, or after a stage or step in the process of genetically engineering the cells for later or subsequent analysis, e.g., such as an analysis after a composition of the engineered cells has been administered to a subject.
  • the freeze and subsequent thaw step removes granulocytes and, to some extent, monocytes in the cell population.
  • the cells are suspended in a freezing solution, e.g., following a washing step to remove plasma and platelets. Any of a variety of known freezing solutions and parameters in some aspects may be used.
  • a freezing solution e.g., following a washing step to remove plasma and platelets.
  • Any of a variety of known freezing solutions and parameters in some aspects may be used.
  • PBS containing 20% DMSO and 8% human serum albumin (HSA), or other suitable cell freezing media. This is then diluted 1: 1 with media so that the final concentration of DMSO and HSA are 10% and 4%, respectively.
  • the cells are generally then frozen to -80° C. at a rate of 1° per minute and stored in the vapor phase of a liquid nitrogen storage tank.
  • recombinant nucleic acids are transferred into cells using recombinant infectious virus particles, such as, e.g., vectors derived from simian virus 40 (SV40), adenoviruses, adeno-associated virus (AAV).
  • recombinant nucleic acids are transferred into T cells using recombinant lentiviral vectors or retroviral vectors, such as gamma-retroviral vectors (see, e.g., Koste et al. (2014) Gene Therapy 2014 Apr 3. doi: 10.1038/gt.2014.25 ; Carlens et al.
  • the virus is adeno-associated virus (AAV).
  • AAV adeno-associated virus
  • the retroviral vector has a long terminal repeat sequence (LTR), e.g., a retroviral vector derived from the Moloney murine leukemia virus (MoMLV), myeloproliferative sarcoma virus (MPSV), murine embryonic stem cell virus (MESV), murine stem cell virus (MSCV), spleen focus forming virus (SFFV).
  • LTR long terminal repeat sequence
  • MoMLV Moloney murine leukemia virus
  • MPSV myeloproliferative sarcoma virus
  • MSV murine embryonic stem cell virus
  • MSCV murine stem cell virus
  • SFFV spleen focus forming virus
  • retroviral vectors are derived from murine retroviruses.
  • the retroviruses include those derived from any avian or mammalian cell source.
  • the retroviruses typically are amphotropic, meaning that they are capable of infecting host cells of several species, including humans.
  • the gene to be expressed replaces the retroviral gag, pol and/or env sequences.
  • retroviral systems e.g., U.S. Pat. Nos. 5,219,740 ; 6,207,453 ; 5,219,740 ; Miller and Rosman (1989) BioTechniques 7:980-990 ; Miller, A. D. (1990) Human Gene Therapy 1:5-14 ; Scarpa et al. (1991) Virology 180:849-852 ; Burns et al. (1993) Proc. Natl. Acad. Sci. USA 90:8033-8037 ; and Boris-Lawrie and Temin (1993) Cur. Opin. Genet. Develop. 3:102-109 .
  • the viral vector particles contain a genome derived from a retroviral genome based vector, such as derived from a lentiviral genome or a gammaretroviral based vector.
  • the heterologous nucleic acid encoding a recombinant receptor, such as an antigen receptor, such as a CAR is contained and/or located between the 5' LTR and 3' LTR sequences of the vector genome.
  • the viral vector genome is a lentivirus genome, such as an HIV-1 genome or an SIV genome.
  • lentiviral vectors have been generated by multiply attenuating virulence genes, for example, the genes env, vif, vpu and nef can be deleted, making the vector safer for therapeutic purposes. Lentiviral vectors are known. See Naldini et al., (1996 and 1998); Zufferey et al., (1997); Dull et al., 1998, U.S. Pat. Nos. 6,013,516 ; and 5,994,136 ).
  • these viral vectors are plasmid-based or virus-based, and are configured to carry the essential sequences for incorporating foreign nucleic acid, for selection, and for transfer of the nucleic acid into a host cell.
  • Known lentiviruses can be readily obtained from depositories or collections such as the American Type Culture Collection ("ATCC"; 10801 University Boulevard., Manassas, Va. 20110-2209), or isolated from known sources using commonly available techniques.
  • Non-limiting examples of lentiviral vectors include those derived from a lentivirus, such as Human Immunodeficiency Virus 1 (HIV-1), HIV-2, an Simian Immunodeficiency Virus (SIV), Human T-lymphotropic virus 1 (HTLV-1), HTLV-2 or equine infection anemia virus (E1AV).
  • lentiviral vectors have been generated by multiply attenuating the HIV virulence genes, for example, the genes env, vif, vpr, vpu and nef are deleted, making the vector safer for therapeutic purposes.
  • Lentiviral vectors are known in the art, see Naldini et al., (1996 and 1998); Zufferey et al., (1997); Dull et al., 1998, U.S. Pat. Nos. 6,013,516 ; and 5,994,136 ).
  • these viral vectors are plasmid-based or virus-based, and are configured to carry the essential sequences for incorporating foreign nucleic acid, for selection, and for transfer of the nucleic acid into a host cell.
  • Known lentiviruses can be readily obtained from depositories or collections such as the American Type Culture Collection ("ATCC"; 10801 University Boulevard., Manassas, Va. 20110-2209), or isolated from known sources using commonly available techniques.
  • ATCC American Type Culture Collection
  • the viral genome vector can contain sequences of the 5' and 3' LTRs of a retrovirus, such as a lentivirus.
  • the viral genome construct may contain sequences from the 5' and 3' LTRs of a lentivirus, and in particular can contain the R and U5 sequences from the 5' LTR of a lentivirus and an inactivated or self-inactivating 3' LTR from a lentivirus.
  • the LTR sequences can be LTR sequences from any lentivirus from any species. For example, they may be LTR sequences from HIV, SIV, FIV or BIV. Typically, the LTR sequences are HIV LTR sequences.
  • the nucleic acid of a viral vector such as an HIV viral vector, lacks additional transcriptional units.
  • the vector genome can contain an inactivated or self-inactivating 3' LTR ( Zufferey et al. J Virol 72: 9873, 1998 ; Miyoshi et al., J Virol 72:8150, 1998 ).
  • deletion in the U3 region of the 3' LTR of the nucleic acid used to produce the viral vector RNA can be used to generate self-inactivating (SIN) vectors. This deletion can then be transferred to the 5' LTR of the proviral DNA during reverse transcription.
  • a self-inactivating vector generally has a deletion of the enhancer and promoter sequences from the 3' long terminal repeat (LTR), which is copied over into the 5' LTR during vector integration. In some instances enough sequence can be eliminated, including the removal of a TATA box, to abolish the transcriptional activity of the LTR. This can prevent production of full-length vector RNA in transduced cells.
  • the U3 element of the 3' LTR contains a deletion of its enhancer sequence, the TATA box, Sp1, and NF-kappa B sites.
  • the self-inactivating 3' LTR can be constructed by any method known in the art. In some instances, this does not affect vector titers or the in vitro or in vivo properties of the vector.
  • the U3 sequence from the lentiviral 5' LTR can be replaced with a promoter sequence in the viral construct, such as a heterologous promoter sequence.
  • a promoter sequence in the viral construct such as a heterologous promoter sequence.
  • An enhancer sequence can also be included. Any enhancer/promoter combination that increases expression of the viral RNA genome in the packaging cell line may be used.
  • the CMV enhancer/promoter sequence is used ( U.S. Pat. No. 5,385,839 and U.S. Pat. No. 5,168,062 ).
  • the risk of insertional mutagenesis can be minimized by constructing the retroviral vector genome, such as lentiviral vector genome, to be integration defective.
  • retroviral vector genome such as lentiviral vector genome
  • a variety of approaches can be pursued to produce a non-integrating vector genome.
  • a mutation(s) can be engineered into the integrase enzyme component of the pol gene, such that it encodes a protein with an inactive integrase.
  • the vector genome itself can be modified to prevent integration by, for example, mutating or deleting one or both attachment sites, or making the 3' LTR-proximal polypurine tract (PPT) non-functional through deletion or modification.
  • PPT 3' LTR-proximal polypurine tract
  • non-genetic approaches are available; these include pharmacological agents that inhibit one or more functions of integrase.
  • the approaches are not mutually exclusive; that is, more than one of them can be used at a time.
  • both the integrase and attachment sites can be non-functional, or the integrase and PPT site can be non-functional, or the attachment sites and PPT site can be non-functional, or all of them can be non-functional.
  • Such methods and viral vector genomes are known and available (see Philpott and Thrasher, Human Gene Therapy 18:483, 2007 ; Engelman et al.
  • the disclosed methods involve methods of transducing cells by contacting, e.g., incubating, a cell composition comprising a plurality of cells with a viral particle.
  • the cells to be transfected or transduced are or comprise primary cells obtained from a subject, such as cells enriched and/or selected from a subject.
  • the concentration of cells to be transduced of the composition is from or from about 1.0 ⁇ 10 5 cells/mL to 1.0 ⁇ 10 8 cells/mL, such as at least or about at least or about 1.0 ⁇ 10 5 cells/mL, 5 ⁇ 10 5 cells/mL, 1 ⁇ 10 6 cells/mL, 5 ⁇ 10 6 cells/mL, 1 ⁇ 10 7 cells/mL, 5 ⁇ 10 7 cells/mL or 1 ⁇ 10 8 cells/mL.
  • the viral particles are provided at a certain ratio of copies of the viral vector particles or infectious units (IU) thereof, per total number of cells to be transduced (IU/cell) .
  • the viral particles are present during the contacting at or about or at least at or about 0.5, 1, 2, 3, 4, 5, 10, 15, 20, 30, 40, 50, or 60 IU of the viral vector particles per one of the cells.
  • the titer of viral vector particles is between or between about 1 ⁇ 10 6 IU/mL and 1 ⁇ 10 8 IU/mL, such as between or between about 5 ⁇ 10 6 IU/mL and 5 ⁇ 10 7 IU/mL, such as at least 6 ⁇ 10 6 IU/mL, 7 ⁇ 10 6 IU/mL, 8 ⁇ 10 6 IU/mL, 9 ⁇ 10 6 IU/mL, 1 ⁇ 10 7 IU/mL, 2 ⁇ 10 7 IU/mL, 3 ⁇ 10 7 IU/mL, 4 ⁇ 10 7 IU/mL, or 5 ⁇ 10 7 IU/mL.
  • transduction can be achieved at a multiplicity of infection (MOI) of less than 100, such as generally less than 60, 50, 40, 30, 20, 10, 5 or less.
  • MOI multiplicity of infection
  • the method involves contacting or incubating, the cells with the viral particles.
  • the contacting is for 30 minutes to 72 hours, such as 30 minute to 48 hours, 30 minutes to 24 hours or 1 hour to 24 hours, such as at least or about at least 30 minutes, 1 hour, 2 hours, 6 hours, 12 hours, 24 hours, 36 hours or more.
  • the composition containing cells, viral particles and reagent can be rotated, generally at relatively low force or speed, such as speed lower than that used to pellet the cells, such as from or from about 600 rpm to 1700 rpm (e.g., at or about or at least 600 rpm, 1000 rpm, or 1500 rpm or 1700 rpm).
  • the rotation is carried at a force, e.g., a relative centrifugal force, of from or from about 100 g to 3200 g (e.g., at or about or at least at or about 100 g, 200 g, 300 g, 400 g, 500 g, 1000 g, 1500 g, 2000 g, 2500 g, 3000 g or 3200 g), as measured for example at an internal or external wall of the chamber or cavity.
  • a force e.g., a relative centrifugal force, of from or from about 100 g to 3200 g (e.g., at or about or at least at or about 100 g, 200 g, 300 g, 400 g, 500 g, 1000 g, 1500 g, 2000 g, 2500 g, 3000 g or 3200 g), as measured for example at an internal or external wall of the chamber or cavity.
  • RCF relative centrifugal force
  • RCF relative centrifugal force
  • the value may be determined using well-known formulas, taking into account the gravitational force, rotation speed and the radius of rotation (distance from the axis of rotation and the object, substance, or particle at which RCF is being measured).
  • the input cells are treated, incubated, or contacted with particles that comprise binding molecules that bind to or recognize the recombinant receptor that is encoded by the viral DNA.
  • recombinant nucleic acids are transferred into T cells via electroporation (see, e.g., Chicaybam et al, (2013) PLoS ONE 8(3): e60298 and Van Tedeloo et al. (2000) Gene Therapy 7(16): 1431-1437 ).
  • recombinant nucleic acids are transferred into T cells via transposition (see, e.g., Manuri et al. (2010) Hum Gene Ther 21(4): 427-437 ; Sharma et al. (2013) Molec Ther Nucl Acids 2, e74 ; and Huang et al. (2009) Methods Mol Biol 506: 115-126 ).
  • transposons suitable for use with mammalian cells, e.g., human primary leukocytes, include but are not limited to Sleeping Beauty and Piggybac.
  • Transposon-based transfection is a two-component system consisting of a transposase and a transposon.
  • the system comprises a transposon is engineered to comprise a foreign DNA (also referred herein as cargo DNA), e.g., a gene encoding a recombinant receptor, that is flanked by inverted repeat/direct repeat (IR/DR) sequences that are recognized by an accompanying tranposase.
  • a foreign DNA also referred herein as cargo DNA
  • IR/DR inverted repeat/direct repeat
  • a non-viral plasmid encodes a transposase under the control of a promoter.
  • transfection of the plasmid into a host cell results in a transitory expression of the transposase at sufficiently levels to integrate the transposon into the genomic DNA.
  • SB Sleeping Beauty
  • IR/DR inverted repeat/direct repeat
  • the SB transposase mediates integration of the transposon, a mobile element encoding a cargo sequence flanked on both sides by inverted terminal repeats that harbor binding sites for the catalytic enzyme (SB). Stable expression results when SB inserts gene sequences into vertebrate chromosomes at a TA target dinucleotide through a cut-and-paste mechanism.
  • This system has been used to engineer a variety of vertebrate cell types, including primary human peripheral blood leukocytes.
  • the cells are contacted, incubated, and/or treated with an SB transposon comprising a cargo gene, e.g., a gene encoding a recombinant receptor or a CAR, flanked by SB IR sequences.
  • the cells to be transfected are contacted, incubated, and/or treated with a plasmid comprising an SB transposon comprising a cargo gene, e.g., a gene encoding a CAR, flanked by SB IR sequences.
  • a plasmid comprising an SB transposon comprising a cargo gene, e.g., a gene encoding a CAR, flanked by SB IR sequences.
  • the plasmid further comprises a gene encoding an SB transposase that is not flanked by SB IR sequences.
  • PiggyBac is another transposon system that can be used to integrate cargo DNA into a host's, e.g., a human's, genomic DNA.
  • the PB transposase recognizes PB transposon-specific inverted terminal repeat sequences (ITRs) located on both ends of the transposon and efficiently moves the contents from the original sites and efficiently integrates them into TTAA chromosomal sites.
  • ITRs inverted terminal repeat sequences
  • the PB transposon system enables genes of interest between the two ITRs in the PB vector to be mobilized into target genomes.
  • the PB system has been used to engineer a variety of vertebrate cell types, including primary human cells.
  • the cells to be transfected are contacted, incubated, and/or treated with an PB transposon comprising a cargo gene, e.g., a gene encoding a CAR, flanked by PB IR sequences.
  • the cells to be transfected are contacted, incubated, and/or treated with a plasmid comprising a PB transposon comprising a cargo gene, e.g., a gene encoding a CAR, flanked by PB IR sequences.
  • the plasmid further comprises a gene encoding an SB transposase that is not flanked by PB IR sequences.
  • transduction with transposons is performed with a plasmid that comprises a transposase gene and a plasmid that comprises a transposon that contains a cargo DNA sequence that is flanked by inverted repeat/direct repeat (IR/DR) sequences that are recognized by the transposase.
  • the cargo DNA sequence encodes a heterologous protein, e.g., a recombinant T cell receptor or a CAR.
  • the plasmid comprises transposase and the transposon.
  • the transposase is under control of a ubiquitous promoter, or any promoter suitable to drive expression of the transposase in the target cell.
  • Ubiquitous promoters include, but are not limited to, EF1a, CMB, SV40, PGK1, Ubc, human ⁇ -actin, CAG, TRE, UAS, Ac5, CaMKIIa, and U6.
  • the cargo DNA comprises a selection cassette allowing for the selection of cells with stable integration of the cargo DNA into the genomic DNA.
  • Suitable selection cassettes include, but are not limited to, selection cassettes encoding a kanamycin resistance gene, spectinomycin resistance gene, streptomycin resistance gene, ampicillin resistance gene, carbenicillin resistance gene, hygromycin resistance gene, bleomycin resistance gene, erythromycin resistance gene, and polymyxin B resistance gene.
  • the components for transduction with a transposon are introduced into the target cell.
  • Any convenient protocol may be employed, where the protocol may provide for in vitro or in vivo introduction of the system components into the target cell, depending on the location of the target cell.
  • the system may be introduced directly into the cell under cell culture conditions permissive of viability of the target cell, e.g., by using standard transformation techniques.
  • Such techniques include, but are not necessarily limited to: viral infection, transformation, conjugation, protoplast fusion, electroporation, particle gun technology, calcium phosphate precipitation, direct microinjection, viral vector delivery, and the like.
  • the SB transposon and the SB transposase source are introduced into a target cell of a multicellular organism, e.g., a mammal or a human, under conditions sufficient for excision of the inverted repeat flanked nucleic acid from the vector carrying the transposon and subsequent integration of the excised nucleic acid into the genome of the target cell.
  • Some instances further comprise a step of ensuring that the requisite transposase activity is present in the target cell along with the introduced transposon.
  • the method may further include introducing a second vector into the target cell which encodes the requisite transposase activity.
  • the amount of vector nucleic acid comprising the transposon and the amount of vector nucleic acid encoding the transposase that is introduced into the cell is sufficient to provide for the desired excision and insertion of the transposon nucleic acid into the target cell genome.
  • the amount of vector nucleic acid introduced should provide for a sufficient amount of transposase activity and a sufficient copy number of the nucleic acid that is desired to be inserted into the target cell.
  • the amount of vector nucleic acid that is introduced into the target cell varies depending on the efficiency of the particular introduction protocol that is employed, e.g., the particular ex vivo administration protocol that is employed.
  • the nucleic acid region of the vector that is flanked by inverted repeats i.e. the vector nucleic acid positioned between the Sleeping Beauty transposase recognized inverted repeats, is excised from the vector via the disclosed transposase and inserted into the genome of the targeted cell.
  • introduction of the vector DNA into the target cell is followed by subsequent transposase mediated excision and insertion of the exogenous nucleic acid carried by the vector into the genome of the targeted cell.
  • the vector is integrated into the genomes of at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6% at least 7% at least 8%, at least 9%, at least 10%, at least 15%, or at least 20% of the cells that are transfected with the SB transposon and/or SB transposase.
  • integration of the nucleic acid into the target cell genome is stable, i.e., the vector nucleic acid remains present in the target cell genome for more than a transient period of time and is passed on a part of the chromosomal genetic material to the progeny of the target cell.
  • the transposons are used to integrate nucleic acids, i.e. polynucleotides, of various sizes into the target cell genome.
  • the size of DNA that is inserted into a target cell genome using the subject methods ranges from about 0.1 kb to 200 kb, from about 0.5 kb to 100 kb, from about 1.0 kb to about 8.0 kb, from about 1.0 to about 200 kb, from about 1.0 to about 10 kb, from about 10 kb to about 50 kb, from about 50 kb to about 100 kb, or from about 100 kb to about 200 kb.
  • the size of DNA that is inserted into a target cell genome using the subject methods ranges from about 1.0 kb to about 8.0 kb. In some instances, the size of DNA that is inserted into a target cell genome using the subject methods ranges from about 1.0 to about 200 kb. In particular instances, the size of DNA that is inserted into a target cell genome using the subject methods ranges from about 1.0 kb to about 8.0 kb.
  • the cells may be transfected either during or after expansion e.g. with a T cell receptor (TCR) or a chimeric antigen receptor (CAR).
  • TCR T cell receptor
  • CAR chimeric antigen receptor
  • This transfection for the introduction of the gene of the desired receptor can be carried out with any suitable retroviral vector, for example.
  • the genetically modified cell population can then be liberated from the initial stimulus (the anti-CD3/anti-CD28 stimulus, for example) and subsequently be stimulated with a second type of stimulus e.g. via a de novo introduced receptor).
  • This second type of stimulus may include an antigenic stimulus in form of a peptide/MHC molecule, the cognate (cross-linking) ligand of the genetically introduced receptor (e.g.
  • a vector may be used that does not require that the cells, e.g., T cells, are activated.
  • the cells may be selected and/or transduced prior to activation.
  • the cells may be engineered prior to, or subsequent to culturing of the cells, and in some cases at the same time as or during at least a portion of the culturing.
  • genes for introduction are those to improve the efficacy of therapy, such as by promoting viability and/or function of transferred cells; genes to provide a genetic marker for selection and/or evaluation of the cells, such as to assess in vivo survival or localization; genes to improve safety, for example, by making the cell susceptible to negative selection in vivo as described by Lupton S. D. et al., Mol. and Cell Biol., 11:6 (1991 ); and Riddell et al., Human Gene Therapy 3:319-338 (1992 ); see also the publications of PCT/US91/08442 and PCT/US94/05601 by Lupton et al.
  • the cells for use in or administered in connection with the disclosed methods contain or are engineered to contain an engineered receptor, e.g., an engineered antigen receptor, such as a chimeric antigen receptor (CAR), or a T cell receptor (TCR).
  • an engineered receptor e.g., an engineered antigen receptor, such as a chimeric antigen receptor (CAR), or a T cell receptor (TCR).
  • populations of such cells compositions containing such cells and/or enriched for such cells, such as in which cells of a certain type such as T cells or CD8+ or CD4+ T cells are enriched or selected.
  • compositions are pharmaceutical compositions and formulations for administration, such as for adoptive cell therapy.
  • methods for administering the cells and compositions to subjects e.g., patients, in accord with the disclosed methods, and/or with the disclosed articles of manufacture or compositions.
  • the cells include one or more nucleic acids introduced via genetic engineering, and thereby express recombinant or genetically engineered products of such nucleic acids.
  • gene transfer is accomplished by first stimulating the cells, such as by combining it with a stimulus that induces a response such as proliferation, survival, and/or activation, e.g., as measured by expression of a cytokine or activation marker, followed by transduction of the activated cells, and expansion in culture to numbers sufficient for clinical applications.
  • the cells generally express recombinant receptors, such as antigen receptors including functional non-TCR antigen receptors, e.g., chimeric antigen receptors (CARs), and other antigen-binding receptors such as transgenic T cell receptors (TCRs). Also among the receptors are other chimeric receptors.
  • antigen receptors including functional non-TCR antigen receptors, e.g., chimeric antigen receptors (CARs), and other antigen-binding receptors such as transgenic T cell receptors (TCRs).
  • CARs chimeric antigen receptors
  • TCRs transgenic T cell receptors
  • chimeric receptors such as a chimeric antigen receptors, contain one or more domains that combine a ligand-binding domain (e.g. antibody or antibody fragment) that provides specificity for a desired antigen (e.g., tumor antigen) with intracellular signaling domains.
  • the intracellular signaling domain is an activating intracellular domain portion, such as a T cell activating domain, providing a primary activation signal.
  • the intracellular signaling domain contains or additionally contains a costimulatory signaling domain to facilitate effector functions.
  • chimeric receptors when genetically engineered into immune cells can modulate T cell activity, and, in some cases, can modulate T cell differentiation or homeostasis, thereby resulting in genetically engineered cells with improved longevity, survival and/or persistence in vivo , such as for use in adoptive cell therapy methods.
  • engineered cells such as T cells
  • a CAR with specificity for a particular antigen (or marker or ligand), such as an antigen expressed on the surface of a particular cell type.
  • the antigen is a polypeptide. In some instances, it is a carbohydrate or other molecule.
  • the antigen is selectively expressed or overexpressed on cells of the disease or condition, e.g., the tumor or pathogenic cells, as compared to normal or non-targeted cells or tissues. In other instances, the antigen is expressed on normal cells and/or is expressed on the engineered cells.
  • the recombinant receptor such as chimeric receptor, contains an intracellular signaling region, which includes a cytoplasmic signaling domain (also interchangeably called an intracellular signaling domain), such as a cytoplasmic (intracellular) region capable of inducing a primary activation signal in a T cell, for example, a cytoplasmic signaling domain of a T cell receptor (TCR) component (e.g. a cytoplasmic signaling domain of a zeta chain of a CD3-zeta (CD3 ⁇ ) chain or a functional variant or signaling portion thereof) and/or that comprises an immunoreceptor tyrosine-based activation motif (ITAM).
  • TCR T cell receptor
  • ITAM immunoreceptor tyrosine-based activation motif
  • the chimeric receptor further contains an extracellular ligand-binding domain that specifically binds to a ligand (e.g. antigen) antigen.
  • the chimeric receptor is a CAR that contains an extracellular antigen-recognition domain that specifically binds to an antigen.
  • the ligand such as an antigen, is a protein expressed on the surface of cells.
  • the CAR is a TCR-like CAR and the antigen is a processed peptide antigen, such as a peptide antigen of an intracellular protein, which, like a TCR, is recognized on the cell surface in the context of a major histocompatibility complex (MHC) molecule.
  • MHC major histocompatibility complex
  • Exemplary antigen receptors including CARs, and methods for engineering and introducing such receptors into cells, include those described, for example, in international patent application publication numbers WO200014257 , WO2013126726 , WO2012/129514 , WO2014031687 , WO2013/166321 , WO2013/071154 , WO2013/123061 U.S. patent application publication numbers US2002131960 , US2013287748 , US20130149337 , U.S.
  • the antigen receptors include a CAR as described in U.S. Patent No.: 7,446,190 , and those described in International Patent Application Publication No.: WO/2014055668 A1 .
  • Examples of the CARs include CARs as disclosed in any of the aforementioned publications, such as WO2014031687 , US 8,339,645 , US 7,446,179 , US 2013/0149337 , U.S.
  • Patent No.: 7,446,190 US Patent No.: 8,389,282 , Kochenderfer et al., 2013, Nature Reviews Clinical Oncology, 10, 267-276 (2013 ); Wang et al. (2012) J. Immunother. 35(9): 689-701 ; and Brentjens et al., Sci Transl Med. 2013 5(177 ). See also WO2014031687 , US 8,339,645 , US 7,446,179 , US 2013/0149337 , U.S. Patent No.: 7,446,190 , and US Patent No.: 8,389,282 .
  • the CAR is constructed with a specificity for a particular antigen (or marker or ligand), such as an antigen expressed in a particular cell type to be targeted by adoptive therapy, e.g., a cancer marker, and/or an antigen intended to induce a dampening response, such as an antigen expressed on a normal or non-diseased cell type.
  • a particular antigen or marker or ligand
  • the CAR typically includes in its extracellular portion one or more antigen binding molecules, such as one or more antigen-binding fragment, domain, or portion, or one or more antibody variable domains, and/or antibody molecules.
  • the CAR includes an antigen-binding portion or portions of an antibody molecule, such as a single-chain antibody fragment (scFv) derived from the variable heavy (VH) and variable light (VL) chains of a monoclonal antibody (mAb).
  • an antibody molecule such as a single-chain antibody fragment (scFv) derived from the variable heavy (VH) and variable light (VL) chains of a monoclonal antibody (mAb).
  • the antibody or antigen-binding portion thereof is expressed on cells as part of a recombinant receptor, such as an antigen receptor.
  • a recombinant receptor such as an antigen receptor.
  • the antigen receptors are functional non-TCR antigen receptors, such as chimeric antigen receptors (CARs).
  • CARs chimeric antigen receptors
  • a CAR containing an antibody or antigen-binding fragment that exhibits TCR-like specificity directed against peptide-MHC complexes also may be referred to as a TCR-like CAR.
  • the extracellular antigen binding domain specific for an MHC-peptide complex of a TCR-like CAR is linked to one or more intracellular signaling components, in some aspects via linkers and/or transmembrane domain(s).
  • such molecules can typically mimic or approximate a signal through a natural antigen receptor, such as a TCR, and, optionally, a signal through such a receptor in combination with a costimulatory receptor
  • the recombinant receptor such as a chimeric receptor (e.g. CAR)
  • a chimeric receptor e.g. CAR
  • the recombinant receptor includes a ligand-binding domain that binds, such as specifically binds, to an antigen (or a ligand).
  • an antigen or a ligand
  • the antigens targeted by the chimeric receptors are those expressed in the context of a disease, condition, or cell type to be targeted via the adoptive cell therapy.
  • diseases and conditions are proliferative, neoplastic, and malignant diseases and disorders, including cancers and tumors, including hematologic cancers, cancers of the immune system, such as lymphomas, leukemias, and/or myelomas, such as B, T, and myeloid leukemias, lymphomas, and multiple myelomas.
  • cancers and tumors including hematologic cancers, cancers of the immune system, such as lymphomas, leukemias, and/or myelomas, such as B, T, and myeloid leukemias, lymphomas, and multiple myelomas.
  • the antigen (or a ligand) is a polypeptide. In some instances, it is a carbohydrate or other molecule. In some instances, the antigen (or a ligand) is selectively expressed or overexpressed on cells of the disease or condition, e.g., the tumor or pathogenic cells, as compared to normal or non-targeted cells or tissues. In other instances, the antigen is expressed on normal cells and/or is expressed on the engineered cells.
  • the CAR contains an antibody or an antigen-binding fragment (e.g. scFv) that specifically recognizes an antigen, such as an intact antigen, expressed on the surface of a cell.
  • an antigen-binding fragment e.g. scFv
  • the antigen (or a ligand) is a tumor antigen or cancer marker.
  • the antigen (or a ligand) is or includes orphan tyrosine kinase receptor (ROR1), B cell maturation antigen (BCMA), carbonic anhydrase 9 (CA9, also known as CAIX or G250), Her2/neu (receptor tyrosine kinase erbB2), CD19, CD20, CD22, mesothelin (MSLN), carcinoembryonic antigen (CEA), and hepatitis B surface antigen, anti-folate receptor, CD23, CD24, CD30, CD33, CD38, CD44, chondroitin sulfate proteoglycan 4 (CSPG4), EGFR, epithelial glycoprotein 2 (EPG-2), epithelial glycoprotein 40 (EPG-40), ephrin receptor A2 (EPHa2), Her3 (erb-B3)
  • ROR1
  • Antigens targeted by the receptors in some instances include antigens associated with a B cell malignancy, such as any of a number of known B cell marker.
  • the antigen targeted by the receptor is CD20, CD19, CD22, ROR1, CD45, CD21, CD5, CD33, Igkappa, Iglambda, CD79a, CD79b or CD30.
  • the antigen is a pathogen-specific antigen.
  • the antigen is a viral antigen (such as a viral antigen from HIV, HCV, HBV, etc.), bacterial antigens, and/or parasitic antigens.
  • the antigen or antigen binding domain is CD19.
  • the scFv contains a VH and a VL derived from an antibody or an antibody fragment specific to CD19.
  • the antibody or antibody fragment that binds CD19 is a mouse derived antibody such as FMC63 and SJ25C1.
  • the antibody or antibody fragment is a human antibody, e.g., as described in U.S. Patent Publication No. US 2016/0152723 .
  • the scFv is derived from FMC63.
  • FMC63 generally refers to a mouse monoclonal IgG1 antibody raised against Nalm-1 and -16 cells expressing CD19 of human origin ( Ling, N. R., et al. (1987). Leucocyte typing III. 302 ).
  • the FMC63 antibody comprises CDRH1 and H2 set forth in SEQ ID NOS: 38, 39 respectively, and CDRH3 set forth in SEQ ID NOS: 40 or 54 and CDRL1 set forth in SEQ ID NOS: 35 and CDR L2 36 or 55 and CDR L3 sequences 37 or 34.
  • the FMC63 antibody comprises the heavy chain variable region (V H ) comprising the amino acid sequence of SEQ ID NO: 41 and the light chain variable region (V L ) comprising the amino acid sequence of SEQ ID NO: 42.
  • the svFv comprises a variable light chain containing the CDRL1 sequence of SEQ ID NO:35, a CDRL2 sequence of SEQ ID NO:36, and a CDRL3 sequence of SEQ ID NO:37 and/or a variable heavy chain containing a CDRH1 sequence of SEQ ID NO:38, a CDRH2 sequence of SEQ ID NO:39, and a CDRH3 sequence of SEQ ID NO:40.
  • the scFv comprises a variable heavy chain region of FMC63 set forth in SEQ ID NO:41 and a variable light chain region of FMC63 set forth in SEQ ID NO:42.
  • the variable heavy and variable light chain are connected by a linker.
  • the linker is set forth in SEQ ID NO:56.
  • the scFv comprises, in order, a V H , a linker, and a V L .
  • the scFv comprises, in order, a V L , a linker, and a V H .
  • the svFc is encoded by a sequence of nucleotides set forth in SEQ ID NO:57 or a sequence that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO:57.
  • the scFv comprises the sequence of amino acids set forth in SEQ ID NO:43 or a sequence that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO:43.
  • the scFv is derived from SJ25C1.
  • SJ25C1 is a mouse monoclonal IgG1 antibody raised against Nalm-1 and -16 cells expressing CD19 of human origin (Ling, N. R., et al. (1987). Leucocyte typing III. 302 ).
  • the SJ25C1 antibody comprises CDRH1, H2 and H3 set forth in SEQ ID NOS: 47-49, respectively, and CDRL1, L2 and L3 sequences set forth in SEQ ID NOS: 44-46, respectively.
  • the SJ25C1 antibody comprises the heavy chain variable region (V H ) comprising the amino acid sequence of SEQ ID NO: 50 and the light chain variable region (V L ) comprising the amino acid sequence of SEQ ID NO: 51.
  • the svFv comprises a variable light chain containing the CDRL1 sequence of SEQ ID NO:44, a CDRL2 sequence of SEQ ID NO: 45, and a CDRL3 sequence of SEQ ID NO:46 and/or a variable heavy chain containing a CDRH1 sequence of SEQ ID NO:47, a CDRH2 sequence of SEQ ID NO:48, and a CDRH3 sequence of SEQ ID NO:49.
  • the scFv comprises a variable heavy chain region of SJ25C1 set forth in SEQ ID NO:50 and a variable light chain region of SJ25C1 set forth in SEQ ID NO:51.
  • the variable heavy and variable light chain are connected by a linker.
  • the linker is set forth in SEQ ID NO:52.
  • the scFv comprises, in order, a V H , a linker, and a V L .
  • the scFv comprises, in order, a V L , a linker, and a V H .
  • the scFv comprises the sequence of amino acids set forth in SEQ ID NO:53 or a sequence that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO:53.
  • the antigen or antigen binding domain is BCMA.
  • the scFv contains a VH and a VL derived from an antibody or an antibody fragment specific to BCMA.
  • the antibody or antibody fragment that binds BCMA is or contains a VH and a VL from an antibody or antibody fragment set forth in International Patent Applications, Publication Number WO 2016/090327 and WO 2016/090320 .
  • the antigen or antigen binding domain is GPRC5D.
  • the scFv contains a VH and a VL derived from an antibody or an antibody fragment specific to GPRC5D.
  • the antibody or antibody fragment that binds GPRC5D is or contains a VH and a VL from an antibody or antibody fragment set forth in International Patent Applications, Publication Number WO 2016/090329 and WO 2016/090312 .
  • the CAR contains a TCR-like antibody, such as an antibody or an antigen-binding fragment (e.g. scFv) that specifically recognizes an intracellular antigen, such as a tumor-associated antigen, presented on the cell surface as a MHC-peptide complex.
  • an antibody or antigen-binding portion thereof that recognizes an MHC-peptide complex can be expressed on cells as part of a recombinant receptor, such as an antigen receptor.
  • the antigen receptors are functional non-TCR antigen receptors, such as chimeric antigen receptors (CARs).
  • CARs chimeric antigen receptors
  • a CAR containing an antibody or antigen-binding fragment that exhibits TCR-like specificity directed against peptide-MHC complexes also may be referred to as a TCR-like CAR.
  • MHC Major histocompatibility complex
  • a protein generally a glycoprotein, that contains a polymorphic peptide binding site or binding groove that can, in some cases, complex with peptide antigens of polypeptides, including peptide antigens processed by the cell machinery.
  • MHC molecules can be displayed or expressed on the cell surface, including as a complex with peptide, i.e. MHC-peptide complex, for presentation of an antigen in a conformation recognizable by an antigen receptor on T cells, such as a TCRs or TCR-like antibody.
  • MHC class I molecules are heterodimers having a membrane spanning ⁇ chain, in some cases with three ⁇ domains, and a non-covalently associated ⁇ 2 microglobulin.
  • MHC class II molecules are composed of two transmembrane glycoproteins, ⁇ and ⁇ , both of which typically span the membrane.
  • An MHC molecule can include an effective portion of an MHC that contains an antigen binding site or sites for binding a peptide and the sequences necessary for recognition by the appropriate antigen receptor.
  • MHC class I molecules deliver peptides originating in the cytosol to the cell surface, where a MHC-peptide complex is recognized by T cells, such as generally CD8 + T cells, but in some cases CD4+ T cells.
  • MHC class II molecules deliver peptides originating in the vesicular system to the cell surface, where they are typically recognized by CD4 + T cells.
  • MHC molecules are encoded by a group of linked loci, which are collectively termed H-2 in the mouse and human leukocyte antigen (HLA) in humans.
  • HLA human leukocyte antigen
  • typically human MHC can also be referred to as human leukocyte antigen (HLA).
  • MHC-peptide complex refers to a complex or association of a peptide antigen and an MHC molecule, such as, generally, by non-covalent interactions of the peptide in the binding groove or cleft of the MHC molecule.
  • the MHC-peptide complex is present or displayed on the surface of cells.
  • the MHC-peptide complex can be specifically recognized by an antigen receptor, such as a TCR, TCR-like CAR or antigen-binding portions thereof.
  • a peptide, such as a peptide antigen or epitope, of a polypeptide can associate with an MHC molecule, such as for recognition by an antigen receptor.
  • the peptide is derived from or based on a fragment of a longer biological molecule, such as a polypeptide or protein.
  • the peptide typically is about 8 to about 24 amino acids in length.
  • a peptide has a length of from or from about 9 to 22 amino acids for recognition in the MHC Class II complex.
  • a peptide has a length of from or from about 8 to 13 amino acids for recognition in the MHC Class I complex.
  • the antigen receptor such as TCR or TCR-like CAR, produces or triggers an activation signal to the T cell that induces a T cell response, such as T cell proliferation, cytokine production, a cytotoxic T cell response or other response.
  • a TCR-like antibody or antigen-binding portion are known or can be produced by methods that are known (see e.g. US Published Application Nos. US 2002/0150914 ; US 2003/0223994 ; US 2004/0191260 ; US 2006/0034850 ; US 2007/00992530 ; US20090226474 ; US20090304679 ; and International PCT Publication No. WO 03/068201 ).
  • an antibody or antigen-binding portion thereof that specifically binds to a MHC-peptide complex can be produced by immunizing a host with an effective amount of an immunogen containing a specific MHC-peptide complex.
  • the peptide of the MHC-peptide complex is an epitope of antigen capable of binding to the MHC, such as a tumor antigen, for example a universal tumor antigen, myeloma antigen or other antigen as described below.
  • an effective amount of the immunogen is then administered to a host for eliciting an immune response, wherein the immunogen retains a three-dimensional form thereof for a period of time sufficient to elicit an immune response against the three-dimensional presentation of the peptide in the binding groove of the MHC molecule.
  • Serum collected from the host is then assayed to determine if desired antibodies that recognize a three-dimensional presentation of the peptide in the binding groove of the MHC molecule is being produced.
  • the produced antibodies can be assessed to confirm that the antibody can differentiate the MHC-peptide complex from the MHC molecule alone, the peptide of interest alone, and a complex of MHC and irrelevant peptide.
  • the desired antibodies can then be isolated.
  • an antibody or antigen-binding portion thereof that specifically binds to an MHC-peptide complex can be produced by employing antibody library display methods, such as phage antibody libraries.
  • phage display libraries of mutant Fab, scFv or other antibody forms can be generated, for example, in which members of the library are mutated at one or more residues of a CDR or CDRs. See e.g. US published application No. US20020150914 , US2014/0294841 ; and Cohen CJ. et al. (2003) J Mol. Recogn. 16:324-332 .
  • antibody herein is used in the broadest sense and includes polyclonal and monoclonal antibodies, including intact antibodies and functional (antigen-binding) antibody fragments, including fragment antigen binding (Fab) fragments, F(ab') 2 fragments, Fab' fragments, Fv fragments, recombinant IgG (rIgG) fragments, variable heavy chain (V H ) regions capable of specifically binding the antigen, single chain antibody fragments, including single chain variable fragments (scFv), and single domain antibodies (e.g. , sdAb, sdFv, nanobody) fragments.
  • Fab fragment antigen binding
  • rIgG fragment antigen binding
  • V H variable heavy chain
  • the term encompasses genetically engineered and/or otherwise modified forms of immunoglobulins, such as intrabodies, peptibodies, chimeric antibodies, fully human antibodies, humanized antibodies, and heteroconjugate antibodies, multispecific, e.g. , bispecific, antibodies, diabodies, triabodies, and tetrabodies, tandem di-scFv, tandem tri-scFv.
  • antibody should be understood to encompass functional antibody fragments thereof.
  • the term also encompasses intact or full-length antibodies, including antibodies of any class or sub-class, including IgG and sub-classes thereof, IgM, IgE, IgA, and IgD.
  • the antigen-binding proteins, antibodies and antigen binding fragments thereof specifically recognize an antigen of a full-length antibody.
  • the heavy and light chains of an antibody can be full-length or can be an antigen-binding portion (a Fab, F(ab')2, Fv or a single chain Fv fragment (scFv)).
  • the antibody heavy chain constant region is chosen from, e.g., IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgD, and IgE, particularly chosen from, e.g., IgG1, IgG2, IgG3, and IgG4, more particularly, IgG1 ( e.g. , human IgG1).
  • the antibody light chain constant region is chosen from, e.g., kappa or lambda, particularly kappa.
  • antibody fragments refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab') 2 ; diabodies; linear antibodies; variable heavy chain (V H ) regions, single-chain antibody molecules such as scFvs and single-domain V H single antibodies; and multispecific antibodies formed from antibody fragments.
  • the antibodies are single-chain antibody fragments comprising a variable heavy chain region and/or a variable light chain region, such as scFvs.
  • variable region refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen.
  • the variable domains of the heavy chain and light chain (V H and V L , respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three CDRs.
  • FRs conserved framework regions
  • a single V H or V L domain may be sufficient to confer antigen-binding specificity.
  • antibodies that bind a particular antigen may be isolated using a V H or V L domain from an antibody that binds the antigen to screen a library of complementary V L or V H domains, respectively. See, e.g. , Portolano et al., J. Immunol. 150:880-887 (1993 ); Clarkson et al., Nature 352:624-628 (1991 ).
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human single-domain antibody.
  • the CAR comprises an antibody heavy chain domain that specifically binds the antigen, such as a cancer marker or cell surface antigen of a cell or disease to be targeted, such as a tumor cell or a cancer cell, such as any of the target antigens described herein or known in the art.
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells.
  • the antibodies are recombinantly-produced fragments, such as fragments comprising arrangements that do not occur naturally, such as those with two or more antibody regions or chains joined by synthetic linkers, e.g. , peptide linkers, and/or that are may not be produced by enzyme digestion of a naturally-occurring intact antibody.
  • the antibody fragments are scFvs.
  • a “humanized” antibody is an antibody in which all or substantially all CDR amino acid residues are derived from non-human CDRs and all or substantially all FR amino acid residues are derived from human FRs.
  • a humanized antibody optionally may include at least a portion of an antibody constant region derived from a human antibody.
  • a "humanized form" of a non-human antibody refers to a variant of the non-human antibody that has undergone humanization, typically to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g. , the antibody from which the CDR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • the chimeric antigen receptor includes an extracellular portion containing an antibody or antibody fragment.
  • the antibody or fragment includes an scFv.
  • the chimeric antigen receptor includes an extracellular portion containing the antibody or fragment and an intracellular signaling region.
  • the intracellular signaling region comprises an intracellular signaling domain.
  • the intracellular signaling domain is or comprises a primary signaling domain, a signaling domain that is capable of inducing a primary activation signal in a T cell, a signaling domain of a T cell receptor (TCR) component, and/or a signaling domain comprising an immunoreceptor tyrosine-based activation motif (ITAM).
  • TCR T cell receptor
  • ITAM immunoreceptor tyrosine-based activation motif
  • the recombinant receptor such as the CAR, such as the antibody portion thereof, further includes a spacer, which may be or include at least a portion of an immunoglobulin constant region or variant or modified version thereof, such as a hinge region, e.g., an IgG4 hinge region, and/or a C H 1/C L and/or Fc region.
  • the recombinant receptor further comprises a spacer and/or a hinge region.
  • the constant region or portion is of a human IgG, such as IgG4 or IgG1.
  • the portion of the constant region serves as a spacer region between the antigen-recognition component, e.g., scFv, and transmembrane domain.
  • the spacer can be of a length that provides for increased responsiveness of the cell following antigen binding, as compared to in the absence of the spacer. In some examples, the spacer is at or about 12 amino acids in length or is no more than 12 amino acids in length.
  • Exemplary spacers include those having at least about 10 to 229 amino acids, about 10 to 200 amino acids, about 10 to 175 amino acids, about 10 to 150 amino acids, about 10 to 125 amino acids, about 10 to 100 amino acids, about 10 to 75 amino acids, about 10 to 50 amino acids, about 10 to 40 amino acids, about 10 to 30 amino acids, about 10 to 20 amino acids, or about 10 to 15 amino acids, and including any integer between the endpoints of any of the listed ranges.
  • a spacer region has about 12 amino acids or less, about 119 amino acids or less, or about 229 amino acids or less.
  • Exemplary spacers include IgG4 hinge alone, IgG4 hinge linked to CH2 and CH3 domains, or IgG4 hinge linked to the CH3 domain.
  • Exemplary spacers include, but are not limited to, those described in Hudecek et al. (2013) Clin. Cancer Res., 19:3153 or international patent application publication number WO2014031687 .
  • the spacer has the sequence set forth in SEQ ID NO: 1, and is encoded by the sequence set forth in SEQ ID NO: 2.
  • the spacer has the sequence set forth in SEQ ID NO: 3.
  • the spacer has the sequence set forth in SEQ ID NO: 4.
  • the constant region or portion is of IgD.
  • the spacer has the sequence set forth in SEQ ID NO: 5.
  • the spacer has a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any of SEQ ID NOS: 1, 3, 4 and 5.
  • the spacer has the sequence set forth in SEQ ID NOS: 23-31.
  • the spacer has a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any of SEQ ID NOS: 23-31.
  • the antigen receptor comprises an intracellular domain linked directly or indirectly to the extracellular domain.
  • the chimeric antigen receptor includes a transmembrane domain linking the extracellular domain and the intracellular signaling domain.
  • the intracellular signaling domain comprises an ITAM.
  • the antigen recognition domain e.g. extracellular domain
  • the chimeric receptor comprises a transmembrane domain linked or fused between the extracellular domain (e.g.
  • the antigen binding component e.g., antibody
  • the transmembrane domain is fused to the extracellular domain.
  • a transmembrane domain that naturally is associated with one of the domains in the receptor e.g., CAR
  • the transmembrane domain is selected or modified by amino acid substitution to avoid binding of such domains to the transmembrane domains of the same or different surface membrane proteins to minimize interactions with other members of the receptor complex.
  • the transmembrane domain in some instances is derived either from a natural or from a synthetic source. Where the source is natural, the domain in some aspects is derived from any membrane-bound or transmembrane protein.
  • Transmembrane regions include those derived from (i.e. comprise at least the transmembrane region(s) of) the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD 16, CD22, CD33, CD37, CD64, CD80, CD86, CD 134, CD137, CD154.
  • the transmembrane domain in some instances is synthetic.
  • the synthetic transmembrane domain comprises predominantly hydrophobic residues such as leucine and valine. In some aspects, a triplet of phenylalanine, tryptophan and valine will be found at each end of a synthetic transmembrane domain. In some instances, the linkage is by linkers, spacers, and/or transmembrane domain(s). In some aspects, the transmembrane domain contains a transmembrane portion of CD28.
  • the extracellular domain and transmembrane domain can be linked directly or indirectly. In some instances, the extracellular domain and transmembrane are linked by a spacer, such as any described herein. In some instances, the receptor contains extracellular portion of the molecule from which the transmembrane domain is derived, such as a CD28 extracellular portion
  • intracellular signaling domains are those that mimic or approximate a signal through a natural antigen receptor, a signal through such a receptor in combination with a costimulatory receptor, and/or a signal through a costimulatory receptor alone.
  • a short oligo- or polypeptide linker for example, a linker of between 2 and 10 amino acids in length, such as one containing glycines and serines, e.g., glycine-serine doublet, is present and forms a linkage between the transmembrane domain and the cytoplasmic signaling domain of the CAR.
  • the receptor e.g., the CAR
  • the receptor generally includes at least one intracellular signaling component or components.
  • the receptor includes an intracellular component of a TCR complex, such as a TCR CD3 chain that mediates T-cell activation and cytotoxicity, e.g., CD3 zeta chain.
  • the ROR1-binding antibody is linked to one or more cell signaling modules.
  • cell signaling modules include CD3 transmembrane domain, CD3 intracellular signaling domains, and/or other CD transmembrane domains.
  • the receptor e.g., CAR, further includes a portion of one or more additional molecules such as Fc receptor ⁇ , CD8, CD4, CD25, or CD16.
  • the CAR includes a chimeric molecule between CD3-zeta (CD3- ⁇ ) or Fc receptor ⁇ and CD8, CD4, CD25 or CD16.
  • the cytoplasmic domain or intracellular signaling domain of the receptor activates at least one of the normal effector functions or responses of the immune cell, e.g., T cell engineered to express the CAR.
  • the CAR induces a function of a T cell such as cytolytic activity or T-helper activity, such as secretion of cytokines or other factors.
  • a truncated portion of an intracellular signaling region of an antigen receptor component or costimulatory molecule is used in place of an intact immunostimulatory chain, for example, if it transduces the effector function signal.
  • the intracellular signaling domain or domains include the cytoplasmic sequences of the T cell receptor (TCR), and in some aspects also those of co-receptors that in the natural context act in concert with such receptor to initiate signal transduction following antigen receptor engagement, and/or any derivative or variant of such molecules, and/or any synthetic sequence that has the same functional capability.
  • TCR T cell receptor
  • full activation In the context of a natural TCR, full activation generally requires not only signaling through the TCR, but also a costimulatory signal. Thus, in some instances, to promote full activation, a component for generating secondary or co-stimulatory signal is also included in the CAR. In other instances, the CAR does not include a component for generating a costimulatory signal. In some aspects, an additional CAR is expressed in the same cell and provides the component for generating the secondary or costimulatory signal.
  • T cell activation is in some aspects described as being mediated by two classes of cytoplasmic signaling sequences: those that initiate antigen-dependent primary activation through the TCR (primary cytoplasmic signaling sequences), and those that act in an antigen-independent manner to provide a secondary or co-stimulatory signal (secondary cytoplasmic signaling sequences).
  • primary cytoplasmic signaling sequences those that initiate antigen-dependent primary activation through the TCR
  • secondary cytoplasmic signaling sequences those that act in an antigen-independent manner to provide a secondary or co-stimulatory signal.
  • the CAR includes one or both of such signaling components.
  • the CAR includes a primary cytoplasmic signaling sequence that regulates primary activation of the TCR complex.
  • Primary cytoplasmic signaling sequences that act in a stimulatory manner may contain signaling motifs which are known as immunoreceptor tyrosine-based activation motifs or ITAMs.
  • ITAM containing primary cytoplasmic signaling sequences include those derived from TCR or CD3 zeta, FcR gamma or FcR beta.
  • cytoplasmic signaling molecule(s) in the CAR contain(s) a cytoplasmic signaling domain, portion thereof, or sequence derived from CD3 zeta.
  • the CAR includes a signaling region and/or transmembrane portion of a costimulatory receptor, such as CD28, 4-1BB, OX40, DAP10, and ICOS. In some aspects, the same CAR includes both the signaling region and costimulatory components.
  • the chimeric antigen receptor contains an intracellular domain derived from a T cell costimulatory molecule or a functional variant thereof, such as between the transmembrane domain and intracellular signaling domain.
  • the T cell costimulatory molecule is CD28 or 41BB.
  • the signaling region and/or activating domain is included within one CAR, whereas the costimulatory component is provided by another CAR recognizing another antigen.
  • the CARs include activating or stimulatory CARs, and costimulatory CARs, both expressed on the same cell (see WO2014/055668 ).
  • the cells include one or more stimulatory or activating CAR and/or a costimulatory CAR.
  • the cells further include inhibitory CARs (iCARs, see Fedorov et al., Sci. Transl.
  • the intracellular signaling domain comprises a CD28 transmembrane and signaling domain linked to a CD3 (e.g., CD3-zeta) intracellular domain.
  • the intracellular signaling domain comprises a chimeric CD28 and CD137 (4-1BB, TNFRSF9) co-stimulatory domains, linked to a CD3 zeta intracellular domain.
  • the CAR encompasses one or more, e.g., two or more, costimulatory domains and an activation domain, e.g., primary activation domain, in the cytoplasmic portion.
  • exemplary CARs include intracellular components of CD3-zeta, CD28, and 4-1BB.
  • the antigen receptor further includes a marker and/or cells expressing the CAR or other antigen receptor further includes a surrogate marker, such as a cell surface marker, which may be used to confirm transduction or engineering of the cell to express the receptor.
  • the marker includes all or part (e.g., truncated form) of CD34, a NGFR, or epidermal growth factor receptor, such as truncated version of such a cell surface receptor (e.g., tEGFR).
  • Exemplary surrogate markers can include truncated forms of cell surface polypeptides, such as truncated forms that are non-functional and to not transduce or are not capable of transducing a signal or a signal ordinarily transduced by the full-length form of the cell surface polypeptide, and/or do not or are not capable of internalizing.
  • Exemplary truncated cell surface polypeptides including truncated forms of growth factors or other receptors such as a truncated human epidermal growth factor receptor 2 (tHER2), a truncated epidermal growth factor receptor (tEGFR, exemplary tEGFR sequence set forth in SEQ ID NO: 7 or 16) or a prostate-specific membrane antigen (PSMA) or modified form thereof.
  • tEGFR may contain an epitope recognized by the antibody cetuximab (Erbitux ® ) or other therapeutic anti-EGFR antibody or binding molecule, which can be used to identify or select cells that have been engineered with the tEGFR construct and an encoded exogenous protein, and/or to eliminate or separate cells expressing the encoded exogenous protein.
  • cetuximab Erbitux ®
  • the marker e.g. surrogate marker, includes all or part ( e.g.
  • CD34 a NGFR
  • CD19 or a truncated CD19 e.g., a truncated non-human CD19
  • epidermal growth factor receptor e.g. , tEGFR
  • the marker is or comprises a fluorescent protein, such as green fluorescent protein (GFP), enhanced green fluorescent protein (EGFP), such as super-fold GFP (sfGFP), red fluorescent protein (RFP), such as tdTomato, mCherry, mStrawberry, AsRed2, DsRed or DsRed2, cyan fluorescent protein (CFP), blue green fluorescent protein (BFP), enhanced blue fluorescent protein (EBFP), and yellow fluorescent protein (YFP), and variants thereof, including species variants, monomeric variants, and codon-optimized and/or enhanced variants of the fluorescent proteins.
  • the marker is or comprises an enzyme, such as a luciferase, the lacZ gene from E.
  • coli alkaline phosphatase, secreted embryonic alkaline phosphatase (SEAP), chloramphenicol acetyl transferase (CAT).
  • exemplary light-emitting reporter genes include luciferase (luc), ⁇ -galactosidase, chloramphenicol acetyltransferase (CAT), ⁇ -glucuronidase (GUS) or variants thereof.
  • the marker is a selection marker.
  • the selection marker is or comprises a polypeptide that confers resistance to exogenous agents or drugs.
  • the selection marker is an antibiotic resistance gene.
  • the selection marker is an antibiotic resistance gene confers antibiotic resistance to a mammalian cell.
  • the selection marker is or comprises a Puromycin resistance gene, a Hygromycin resistance gene, a Blasticidin resistance gene, a Neomycin resistance gene, a Geneticin resistance gene or a Zeocin resistance gene or a modified form thereof.
  • the nucleic acid encoding the marker is operably linked to a polynucleotide encoding for a linker sequence, such as a cleavable linker sequence, e.g. , a T2A.
  • a marker, and optionally a linker sequence can be any as disclosed in PCT Pub. No. WO2014031687 .
  • the marker can be a truncated EGFR (tEGFR) that is, optionally, linked to a linker sequence, such as a T2A cleavable linker sequence.
  • tEGFR truncated EGFR
  • An exemplary polypeptide for a truncated EGFR e.g.
  • tEGFR comprises the sequence of amino acids set forth in SEQ ID NO: 7 or 16 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 7 or 16.
  • the nucleic acid encoding the marker is operably linked to a polynucleotide encoding for a linker sequence, such as a cleavable linker sequence, e.g., MMP cleavable linker sequence.
  • a linker sequence such as a cleavable linker sequence, e.g., MMP cleavable linker sequence.
  • a marker, and optionally a linker sequence can be any as disclosed in published patent application No. WO2014031687 .
  • the marker can be a truncated EGFR (tEGFR) that is, optionally, linked to a linker sequence, such as a T2A cleavable linker sequence.
  • tEGFR truncated EGFR
  • the marker is a molecule, e.g., cell surface protein, not naturally found on T cells or not naturally found on the surface of T cells, or a portion thereof.
  • the molecule is a non-self molecule, e.g., non-self protein, i.e., one that is not recognized as "self' by the immune system of the host into which the cells will be adoptively transferred.
  • the marker serves no therapeutic function and/or produces no effect other than to be used as a marker for genetic engineering, e.g., for selecting cells successfully engineered.
  • the marker may be a therapeutic molecule or molecule otherwise exerting some desired effect, such as a ligand for a cell to be encountered in vivo, such as a costimulatory or immune checkpoint molecule to enhance and/or dampen responses of the cells upon adoptive transfer and encounter with ligand.
  • CARs are referred to as first, second, and/or third generation CARs.
  • a first generation CAR is one that solely provides a CD3-chain induced signal upon antigen binding;
  • a second-generation CARs is one that provides such a signal and costimulatory signal, such as one including an intracellular signaling domain from a costimulatory receptor such as CD28 or CD137;
  • a third generation CAR in some aspects is one that includes multiple costimulatory domains of different costimulatory receptors.
  • the chimeric antigen receptor includes an extracellular portion containing the antibody or fragment described herein. In some aspects, the chimeric antigen receptor includes an extracellular portion containing the antibody or fragment described herein and an intracellular signaling domain. In some instances, the antibody or fragment includes an scFv or a single-domain V H antibody and the intracellular domain contains an ITAM. In some aspects, the intracellular signaling domain includes a signaling domain of a zeta chain of a CD3-zeta (CD3 ⁇ ) chain. In some instances, the chimeric antigen receptor includes a transmembrane domain disposed between the extracellular domain and the intracellular signaling region.
  • the transmembrane domain contains a transmembrane portion of CD28.
  • the extracellular domain and transmembrane can be linked directly or indirectly.
  • the extracellular domain and transmembrane are linked by a spacer, such as any described herein.
  • the chimeric antigen receptor contains an intracellular domain of a T cell costimulatory molecule, such as between the transmembrane domain and intracellular signaling domain.
  • the T cell costimulatory molecule is CD28 or 4-1BB.
  • the CAR contains an antibody, e.g., an antibody fragment, a transmembrane domain that is or contains a transmembrane portion of CD28 or a functional variant thereof, and an intracellular signaling domain containing a signaling portion of CD28 or functional variant thereof and a signaling portion of CD3 zeta or functional variant thereof.
  • the CAR contains an antibody, e.g., antibody fragment, a transmembrane domain that is or contains a transmembrane portion of CD28 or a functional variant thereof, and an intracellular signaling domain containing a signaling portion of a 4-1BB or functional variant thereof and a signaling portion of CD3 zeta or functional variant thereof.
  • the receptor further includes a spacer containing a portion of an Ig molecule, such as a human Ig molecule, such as an Ig hinge, e.g. an IgG4 hinge, such as a hinge-only spacer.
  • an Ig molecule such as a human Ig molecule
  • an Ig hinge e.g. an IgG4 hinge, such as a hinge-only spacer.
  • the transmembrane domain of the receptor e.g., the CAR is a transmembrane domain of human CD28 or variant thereof, e.g., a 27-amino acid transmembrane domain of a human CD28 (Accession No.: P10747.1), or is a transmembrane domain that comprises the sequence of amino acids set forth in SEQ ID NO: 8 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO:8; in some instances, the transmembrane-domain containing portion of the recombinant receptor comprises the sequence of amino acids set forth in SEQ ID NO: 9 or a sequence of amino acids having at least at or about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%,
  • the chimeric antigen receptor contains an intracellular domain of a T cell costimulatory molecule.
  • the T cell costimulatory molecule is CD28 or 4-1BB.
  • the intracellular signaling region comprises an intracellular costimulatory signaling domain of human CD28 or functional variant or portion thereof, such as a 41 amino acid domain thereof and/or such a domain with an LL to GG substitution at positions 186-187 of a native CD28 protein.
  • the intracellular signaling domain can comprise the sequence of amino acids set forth in SEQ ID NO: 10 or 11 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 10 or 11.
  • the intracellular region comprises an intracellular costimulatory signaling domain of 4-1BB or functional variant or portion thereof, such as a 42-amino acid cytoplasmic domain of a human 4-1BB (Accession No. Q07011.1) or functional variant or portion thereof, such as the sequence of amino acids set forth in SEQ ID NO: 12 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 12.
  • 4-1BB intracellular costimulatory signaling domain of 4-1BB or functional variant or portion thereof, such as a 42-amino acid cytoplasmic domain of a human 4-1BB (Accession No. Q07011.1) or functional variant or portion thereof, such as the sequence of amino acids set forth in SEQ ID NO: 12 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 8
  • the intracellular signaling region comprises a human CD3 chain, optionally a CD3 zeta stimulatory signaling domain or functional variant thereof, such as an 112 AA cytoplasmic domain of isoform 3 of human CD3 ⁇ (Accession No.: P20963.2) or a CD3 zeta signaling domain as described in U.S. Patent No.: 7,446,190 or U.S. Patent No. 8,911,993 .
  • the intracellular signaling region comprises the sequence of amino acids set forth in SEQ ID NO: 13, 14 or 15 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 13, 14 or 15.
  • the spacer contains only a hinge region of an IgG, such as only a hinge of IgG4 or IgG1, such as the hinge only spacer set forth in SEQ ID NO:1.
  • the spacer is an Ig hinge, e.g., and IgG4 hinge, linked to a C H 2 and/or C H 3 domains.
  • the spacer is an Ig hinge, e.g., an IgG4 hinge, linked to C H 2 and C H 3 domains, such as set forth in SEQ ID NO:3.
  • the spacer is an Ig hinge, e.g., an IgG4 hinge, linked to a C H 3 domain only, such as set forth in SEQ ID NO:4.
  • the spacer is or comprises a glycine-serine rich sequence or other flexible linker such as known flexible linkers.
  • the CAR includes an antibody such as an antibody fragment, including scFvs, a spacer, such as a spacer containing a portion of an immunoglobulin molecule, such as a hinge region and/or one or more constant regions of a heavy chain molecule, such as an Ig-hinge containing spacer, a transmembrane domain containing all or a portion of a CD28-derived transmembrane domain, a CD28-derived intracellular signaling domain, and a CD3 zeta signaling domain.
  • an antibody such as an antibody fragment, including scFvs
  • a spacer such as a spacer containing a portion of an immunoglobulin molecule, such as a hinge region and/or one or more constant regions of a heavy chain molecule, such as an Ig-hinge containing spacer
  • a transmembrane domain containing all or a portion of a CD28-derived transmembrane domain
  • the CAR includes an antibody or fragment, such as scFv, a spacer such as any of the Ig-hinge containing spacers, a CD28-derived transmembrane domain, a 4-1BB-derived intracellular signaling domain, and a CD3 zeta-derived signaling domain.
  • nucleic acid molecules encoding such CAR constructs further includes a sequence encoding a T2A ribosomal skip element and/or a tEGFR sequence, e.g., downstream of the sequence encoding the CAR.
  • the sequence encodes a T2A ribosomal skip element set forth in SEQ ID NO: 6 or 17, or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 6 or 17.
  • T cells expressing an antigen receptor e.g.
  • CAR can also be generated to express a truncated EGFR (EGFRt) as a non-immunogenic selection epitope (e.g. by introduction of a construct encoding the CAR and EGFRt separated by a T2A ribosome switch to express two proteins from the same construct), which then can be used as a marker to detect such cells (see e.g. U.S. Patent No. 8,802,374 ).
  • EGFRt truncated EGFR
  • the sequence encodes an tEGFR sequence set forth in SEQ ID NO: 7 or 16, or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 7 or 16.
  • the peptide such as T2A, can cause the ribosome to skip (ribosome skipping) synthesis of a peptide bond at the C-terminus of a 2A element, leading to separation between the end of the 2A sequence and the next peptide downstream ( see , for example, de Felipe. Genetic Vaccines and Ther.
  • 2A sequences that can be used in the methods and nucleic acids disclosed herein, without limitation, 2A sequences from the foot-and-mouth disease virus (F2A, e.g., SEQ ID NO: 22), equine rhinitis A virus (E2A, e.g., SEQ ID NO: 21), Thosea asigna virus (T2A, e.g., SEQ ID NO: 6 or 17), and porcine teschovirus-1 (P2A, e.g. , SEQ ID NO: 19 or 20) as described in U.S. Patent Publication No. 20070116690 .
  • F2A foot-and-mouth disease virus
  • E2A equine rhinitis A virus
  • T2A e.g., SEQ ID NO: 6 or 17
  • P2A porcine teschovirus-1
  • the recombinant receptors, such as CARs, expressed by the cells administered to the subject generally recognize or specifically bind to a molecule that is expressed in, associated with, and/or specific for the disease or condition or cells thereof being treated.
  • the receptor Upon specific binding to the molecule, e.g., antigen, the receptor generally delivers an immunostimulatory signal, such as an ITAM-transduced signal, into the cell, thereby promoting an immune response targeted to the disease or condition.
  • the cells express a CAR that specifically binds to an antigen expressed by a cell or tissue of the disease or condition or associated with the disease or condition.
  • CAARs Chimeric Auto-Antibody Receptors
  • CAAR chimeric autoantibody receptor
  • the CAAR is specific for an autoantibody.
  • a cell expressing the CAAR such as a T cell engineered to express a CAAR, can be used to specifically bind to and kill autoantibody-expressing cells, but not normal antibody expressing cells.
  • CAAR-expressing cells can be used to treat an autoimmune disease associated with expression of self-antigens, such as autoimmune diseases.
  • CAAR-expressing cells can target B cells that ultimately produce the autoantibodies and display the autoantibodies on their cell surfaces, mark these B cells as disease-specific targets for therapeutic intervention.
  • CAAR-expressing cells can be used to efficiently targeting and killing the pathogenic B cells in autoimmune diseases by targeting the disease-causing B cells using an antigen-specific chimeric autoantibody receptor.
  • the recombinant receptor is a CAAR, such as any described in U.S. Patent Application Pub. No. US 2017/0051035 .
  • the CAAR comprises an autoantibody binding domain, a transmembrane domain, and an intracellular signaling region.
  • the intracellular signaling region comprises an intracellular signaling domain.
  • the intracellular signaling domain is or comprises a primary signaling domain, a signaling domain that is capable of inducing a primary activation signal in a T cell, a signaling domain of a T cell receptor (TCR) component, and/or a signaling domain comprising an immunoreceptor tyrosine-based activation motif (ITAM).
  • TCR T cell receptor
  • ITAM immunoreceptor tyrosine-based activation motif
  • the intracellular signaling region comprises a secondary or costimulatory signaling region (secondary intracellular signaling regions).
  • the autoantibody binding domain comprises an autoantigen or a fragment thereof.
  • the choice of autoantigen can depend upon the type of autoantibody being targeted.
  • the autoantigen may be chosen because it recognizes an autoantibody on a target cell, such as a B cell, associated with a particular disease state, e.g. an autoimmune disease, such as an autoantibody-mediated autoimmune disease.
  • the autoimmune disease includes pemphigus vulgaris (PV).
  • Exemplary autoantigens include desmoglein 1 (Dsg1) and Dsg3.
  • the cells used in connection with the disclosed methods, uses, articles of manufacture and compositions include cells employing multi-targeting strategies.
  • the cells express multi-chain chimeric antigen receptors (CAR) or express two or more genetically engineered receptors on the cell, each recognizing the same of a different antigen and typically each including a different intracellular signaling component.
  • CAR multi-chain chimeric antigen receptors
  • Such multi-targeting strategies are described, for example, in International Patent Application, Publication No.: WO 2014055668 A1 (describing combinations of activating and costimulatory CARs, e.g., targeting two different antigens present individually on off-target, e.g., normal cells, but present together only on cells of the disease or condition to be treated) and Fedorov et al., Sci.
  • Transl. Medicine, 5(215) (2013 ) (describing cells expressing an activating and an inhibitory CAR, such as those in which the activating CAR binds to one antigen expressed on both normal or non-diseased cells and cells of the disease or condition to be treated, and the inhibitory CAR binds to another antigen expressed only on the normal cells or cells which it is not desired to treat).
  • the cells include a receptor expressing a first genetically engineered antigen receptor (e.g., CAR or TCR) which is capable of inducing an activating or stimulatory signal to the cell, generally upon specific binding to the antigen recognized by the first receptor, e.g., the first antigen.
  • the cell further includes a second genetically engineered antigen receptor (e.g., CAR or TCR), e.g., a chimeric costimulatory receptor, which is capable of inducing a costimulatory signal to the immune cell, generally upon specific binding to a second antigen recognized by the second receptor.
  • the first antigen and second antigen are the same. In some instances, the first antigen and second antigen are different.
  • the first and/or second genetically engineered antigen receptor is capable of inducing an activating signal to the cell.
  • the receptor includes an intracellular signaling component containing ITAM or ITAM-like motifs.
  • the activation induced by the first receptor involves a signal transduction or change in protein expression in the cell resulting in initiation of an immune response, such as ITAM phosphorylation and/or initiation of ITAM-mediated signal transduction cascade, formation of an immunological synapse and/or clustering of molecules near the bound receptor (e.g. CD4 or CD8, etc.), activation of one or more transcription factors, such as NF- ⁇ B and/or AP-1, and/or induction of gene expression of factors such as cytokines, proliferation, and/or survival.
  • an immune response such as ITAM phosphorylation and/or initiation of ITAM-mediated signal transduction cascade
  • formation of an immunological synapse and/or clustering of molecules near the bound receptor e.g. CD4 or CD8, etc.
  • the first and/or second receptor includes intracellular signaling domains or regions of costimulatory receptors such as CD28, CD 137 (4-1BB), OX40, and/or ICOS.
  • the first and second receptor include an intracellular signaling domain of a costimulatory receptor that are different.
  • the first receptor contains a CD28 costimulatory signaling region and the second receptor contain a 4-1BB co-stimulatory signaling region or vice versa.
  • the first and/or second receptor includes both an intracellular signaling domain containing ITAM or ITAM-like motifs and an intracellular signaling domain of a costimulatory receptor.
  • the first receptor contains an intracellular signaling domain containing ITAM or ITAM-like motifs and the second receptor contains an intracellular signaling domain of a costimulatory receptor.
  • the costimulatory signal in combination with the activating signal induced in the same cell is one that results in an immune response, such as a robust and sustained immune response, such as increased gene expression, secretion of cytokines and other factors, and T cell mediated effector functions such as cell killing.
  • neither ligation of the first receptor alone nor ligation of the second receptor alone induces a robust immune response.
  • the cell becomes tolerized or unresponsive to antigen, or inhibited, and/or is not induced to proliferate or secrete factors or carry out effector functions.
  • a desired response is achieved, such as full immune activation or stimulation, e.g., as indicated by secretion of one or more cytokine, proliferation, persistence, and/or carrying out an immune effector function such as cytotoxic killing of a target cell.
  • the two receptors induce, respectively, an activating and an inhibitory signal to the cell, such that binding by one of the receptor to its antigen activates the cell or induces a response, but binding by the second inhibitory receptor to its antigen induces a signal that suppresses or dampens that response.
  • activating CARs and inhibitory CARs or iCARs are combinations of activating CARs and inhibitory CARs or iCARs.
  • Such a strategy may be used, for example, in which the activating CAR binds an antigen expressed in a disease or condition but which is also expressed on normal cells, and the inhibitory receptor binds to a separate antigen which is expressed on the normal cells but not cells of the disease or condition.
  • the multi-targeting strategy is employed in a case where an antigen associated with a particular disease or condition is expressed on a non-diseased cell and/or is expressed on the engineered cell itself, either transiently (e.g., upon stimulation in association with genetic engineering) or permanently.
  • an antigen associated with a particular disease or condition is expressed on a non-diseased cell and/or is expressed on the engineered cell itself, either transiently (e.g., upon stimulation in association with genetic engineering) or permanently.
  • specificity, selectivity, and/or efficacy may be improved.
  • the plurality of antigens are expressed on the cell, tissue, or disease or condition being targeted, such as on the cancer cell.
  • the cell, tissue, disease or condition is multiple myeloma or a multiple myeloma cell.
  • one or more of the plurality of antigens generally also is expressed on a cell which it is not desired to target with the cell therapy, such as a normal or non-diseased cell or tissue, and/or the engineered cells themselves. In such instances, by requiring ligation of multiple receptors to achieve a response of the cell, specificity and/or efficacy is achieved.
  • TCRs T Cell Receptors
  • engineered cells such as T cells
  • TCR T cell receptor
  • a target polypeptide such as an antigen of a tumor, viral or autoimmune protein.
  • a "T cell receptor” or “TCR” is a molecule that contains a variable ⁇ and ⁇ chains (also known as TCR ⁇ and TCR ⁇ , respectively) or a variable ⁇ and ⁇ chains (also known as TCR ⁇ and TCR ⁇ , respectively), or antigen-binding portions thereof, and which is capable of specifically binding to a peptide bound to an MHC molecule.
  • the TCR is in the ⁇ form.
  • TCRs that exist in ⁇ and ⁇ forms are generally structurally similar, but T cells expressing them may have distinct anatomical locations or functions.
  • a TCR can be found on the surface of a cell or in soluble form.
  • a TCR is found on the surface of T cells (or T lymphocytes) where it is generally responsible for recognizing antigens bound to major histocompatibility complex (MHC) molecules.
  • MHC major histocompatibility complex
  • TCR should be understood to encompass full TCRs as well as antigen-binding portions or antigen-binding fragments thereof.
  • the TCR is an intact or full-length TCR, including TCRs in the ⁇ form or ⁇ form.
  • the TCR is an antigen-binding portion that is less than a full-length TCR but that binds to a specific peptide bound in an MHC molecule, such as binds to an MHC-peptide complex.
  • an antigen-binding portion or fragment of a TCR can contain only a portion of the structural domains of a full-length or intact TCR, but yet is able to bind the peptide epitope, such as MHC-peptide complex, to which the full TCR binds.
  • an antigen-binding portion contains the variable domains of a TCR, such as variable ⁇ chain and variable ⁇ chain of a TCR, sufficient to form a binding site for binding to a specific MHC-peptide complex.
  • the variable chains of a TCR contain complementarity determining regions involved in recognition of the peptide, MHC and/or MHC-peptide complex.
  • variable domains of the TCR contain hypervariable loops, or complementarity determining regions (CDRs), which generally are the primary contributors to antigen recognition and binding capabilities and specificity.
  • CDRs complementarity determining regions
  • a CDR of a TCR or combination thereof forms all or substantially all of the antigen-binding site of a given TCR molecule.
  • the various CDRs within a variable region of a TCR chain generally are separated by framework regions (FRs), which generally display less variability among TCR molecules as compared to the CDRs (see, e.g., Jores et al., Proc. Nat'l Acad. Sci. U.S.A. 87:9138, 1990 ; Chothia et al., EMBO J.
  • CDR3 is the main CDR responsible for antigen binding or specificity, or is the most important among the three CDRs on a given TCR variable region for antigen recognition, and/or for interaction with the processed peptide portion of the peptide-MHC complex.
  • the CDR1 of the alpha chain can interact with the N-terminal part of certain antigenic peptides.
  • CDR1 of the beta chain can interact with the C-terminal part of the peptide.
  • CDR2 contributes most strongly to or is the primary CDR responsible for the interaction with or recognition of the MHC portion of the MHC-peptide complex.
  • the variable region of the ⁇ -chain can contain a further hypervariable region (CDR4 or HVR4), which generally is involved in superantigen binding and not antigen recognition ( Kotb (1995) Clinical Microbiology Reviews, 8:411-426 ).
  • a TCR also can contain a constant domain, a transmembrane domain and/or a short cytoplasmic tail (see, e.g., Janeway et al., Immunobiology: The Immune System in Health and Disease, 3rd Ed., Current Biology Publications, p. 4:33, 1997 ).
  • each chain of the TCR can possess one N-terminal immunoglobulin variable domain, one immunoglobulin constant domain, a transmembrane region, and a short cytoplasmic tail at the C-terminal end.
  • a TCR is associated with invariant proteins of the CD3 complex involved in mediating signal transduction.
  • a TCR chain contains one or more constant domain.
  • the extracellular portion of a given TCR chain e.g., ⁇ -chain or ⁇ -chain
  • a constant domain e.g., ⁇ -chain constant domain or C ⁇ , typically positions 117 to 259 of the chain based on Kabat numbering or ⁇ chain constant domain or C ⁇ , typically positions 117 to 295 of the chain based on Kabat
  • the extracellular portion of the TCR formed by the two chains contains two membrane-proximal constant domains, and two membrane-distal variable domains, which variable domains each contain CDRs.
  • the constant domain of the TCR may contain short connecting sequences in which a cysteine residue forms a disulfide bond, thereby linking the two chains of the TCR.
  • a TCR may have an additional cysteine residue in each of the ⁇ and ⁇ chains, such that the TCR contains two disulfide bonds in the constant domains.
  • the TCR chains contain a transmembrane domain. In some instances, the transmembrane domain is positively charged. In some cases, the TCR chain contains a cytoplasmic tail. In some cases, the structure allows the TCR to associate with other molecules like CD3 and subunits thereof. For example, a TCR containing constant domains with a transmembrane region may anchor the protein in the cell membrane and associate with invariant subunits of the CD3 signaling apparatus or complex.
  • the intracellular tails of CD3 signaling subunits e.g. CD3 ⁇ , CD3 ⁇ , CD3 ⁇ and CD3 ⁇ chains
  • the TCR may be a heterodimer of two chains ⁇ and ⁇ (or optionally ⁇ and ⁇ ) or it may be a single chain TCR construct. In some instances, the TCR is a heterodimer containing two separate chains ( ⁇ and ⁇ chains or ⁇ and ⁇ chains) that are linked, such as by a disulfide bond or disulfide bonds.
  • the TCR can be generated from a known TCR sequence(s), such as sequences of V ⁇ , ⁇ chains, for which a substantially full-length coding sequence is readily available. Methods for obtaining full-length TCR sequences, including V chain sequences, from cell sources are well known.
  • nucleic acids encoding the TCR can be obtained from a variety of sources, such as by polymerase chain reaction (PCR) amplification of TCR-encoding nucleic acids within or isolated from a given cell or cells, or synthesis of publicly available TCR DNA sequences.
  • PCR polymerase chain reaction
  • the TCR is obtained from a biological source, such as from cells such as from a T cell (e.g. cytotoxic T cell), T-cell hybridomas or other publicly available source.
  • the T-cells can be obtained from in vivo isolated cells.
  • the TCR is a thymically selected TCR.
  • the TCR is a neoepitope-restricted TCR.
  • the T- cells can be a cultured T-cell hybridoma or clone.
  • the TCR or antigen-binding portion thereof or antigen-binding fragment thereof can be synthetically generated from knowledge of the sequence of the TCR.
  • the TCR is generated from a TCR identified or selected from screening a library of candidate TCRs against a target polypeptide antigen, or target T cell epitope thereof.
  • TCR libraries can be generated by amplification of the repertoire of V ⁇ and V ⁇ from T cells isolated from a subject, including cells present in PBMCs, spleen or other lymphoid organ.
  • T cells can be amplified from tumor-infiltrating lymphocytes (TILs).
  • TCR libraries can be generated from CD4+ or CD8+ T cells.
  • the TCRs can be amplified from a T cell source of a normal of healthy subject, i.e. normal TCR libraries.
  • the TCRs can be amplified from a T cell source of a diseased subject, i.e. diseased TCR libraries.
  • degenerate primers are used to amplify the gene repertoire of V ⁇ and V ⁇ , such as by RT-PCR in samples, such as T cells, obtained from humans.
  • scTv libraries can be assembled from na ⁇ ve V ⁇ and V ⁇ libraries in which the amplified products are cloned or assembled to be separated by a linker.
  • the libraries can be HLA allele-specific.
  • TCR libraries can be generated by mutagenesis or diversification of a parent or scaffold TCR molecule.
  • the TCRs are subjected to directed evolution, such as by mutagenesis, e.g., of the ⁇ or ⁇ chain. In some aspects, particular residues within CDRs of the TCR are altered. In some instances, selected TCRs can be modified by affinity maturation. In some instances, antigen-specific T cells may be selected, such as by screening to assess CTL activity against the peptide. In some aspects, TCRs, e.g. present on the antigen-specific T cells, may be selected, such as by binding activity, e.g., particular affinity or avidity for the antigen.
  • the TCR or antigen-binding portion thereof is one that has been modified or engineered.
  • directed evolution methods are used to generate TCRs with altered properties, such as with higher affinity for a specific MHC-peptide complex.
  • directed evolution is achieved by display methods including, but not limited to, yeast display ( Holler et al. (2003) Nat Immunol, 4, 55-62 ; Holler et al. (2000) Proc Natl Acad Sci USA, 97, 5387-92 ), phage display ( Li et al. (2005) Nat Biotechnol, 23, 349-54 ), or T cell display ( Chervin et al. (2008) J Immunol Methods, 339, 175-84 ).
  • display approaches involve engineering, or modifying, a known, parent or reference TCR.
  • a wild-type TCR can be used as a template for producing mutagenized TCRs in which in one or more residues of the CDRs are mutated, and mutants with an desired altered property, such as higher affinity for a desired target antigen, are selected.
  • peptides of a target polypeptide for use in producing or generating a TCR of interest are known or can be readily identified.
  • peptides suitable for use in generating TCRs or antigen-binding portions can be determined based on the presence of an HLA-restricted motif in a target polypeptide of interest, such as a target polypeptide described below.
  • peptides are identified using available computer prediction models. In some instances, for predicting MHC class I binding sites, such models include, but are not limited to, ProPred1 ( Singh and Raghava (2001) Bioinformatics 17(12): 1236-1237 , and SYFPEITHI (see Schuler et al.
  • the MHC-restricted epitope is HLA-A0201, which is expressed in approximately 39-46% of all Caucasians and therefore, represents a suitable choice of MHC antigen for use preparing a TCR or other MHC-peptide binding molecule.
  • HLA-A0201-binding motifs and the cleavage sites for proteasomes and immune-proteasomes using computer prediction models are known.
  • such models include, but are not limited to, ProPred1 (described in more detail in Singh and Raghava, ProPred: prediction of HLA-DR binding sites. BIOINFORMATICS 17(12):1236-1237 2001 ), and SYFPEITHI (see Schuler et al. SYFPEITHI, Database for Searching and T-Cell Epitope Prediction. in Immunoinformatics Methods in Molecular Biology, vol 409(1): 75-93 2007 )
  • the TCR or antigen binding portion thereof may be a recombinantly produced natural protein or mutated form thereof in which one or more property, such as binding characteristic, has been altered.
  • a TCR may be derived from one of various animal species, such as human, mouse, rat, or other mammal.
  • a TCR may be cell-bound or in soluble form. In some instances, for purposes of the disclosed methods, the TCR is in cell-bound form expressed on the surface of a cell.
  • the TCR is a full-length TCR. In some instances, the TCR is an antigen-binding portion. In some instances, the TCR is a dimeric TCR (dTCR). In some instances, the TCR is a single-chain TCR (sc-TCR). In some instances, a dTCR or scTCR have the structures as described in WO 03/020763 , WO 04/033685 , WO 2011/044186 .
  • the TCR contains a sequence corresponding to the transmembrane sequence. In some instances, the TCR does contain a sequence corresponding to cytoplasmic sequences. In some instances, the TCR is capable of forming a TCR complex with CD3. In some instances, any of the TCRs, including a dTCR or scTCR, can be linked to signaling domains that yield an active TCR on the surface of a T cell. In some instances, the TCR is expressed on the surface of cells.
  • a dTCR contains a first polypeptide wherein a sequence corresponding to a TCR ⁇ chain variable region sequence is fused to the N terminus of a sequence corresponding to a TCR ⁇ chain constant region extracellular sequence, and a second polypeptide wherein a sequence corresponding to a TCR ⁇ chain variable region sequence is fused to the N terminus a sequence corresponding to a TCR ⁇ chain constant region extracellular sequence, the first and second polypeptides being linked by a disulfide bond.
  • the bond can correspond to the native inter-chain disulfide bond present in native dimeric ⁇ TCRs. In some instances, the interchain disulfide bonds are not present in a native TCR.
  • one or more cysteines can be incorporated into the constant region extracellular sequences of dTCR polypeptide pair.
  • both a native and a non-native disulfide bond may be desirable.
  • the TCR contains a transmembrane sequence to anchor to the membrane.
  • a dTCR contains a TCR ⁇ chain containing a variable ⁇ domain, a constant ⁇ domain and a first dimerization motif attached to the C-terminus of the constant ⁇ domain, and a TCR ⁇ chain comprising a variable ⁇ domain, a constant ⁇ domain and a first dimerization motif attached to the C-terminus of the constant ⁇ domain, wherein the first and second dimerization motifs easily interact to form a covalent bond between an amino acid in the first dimerization motif and an amino acid in the second dimerization motif linking the TCR ⁇ chain and TCR ⁇ chain together.
  • the TCR is a scTCR.
  • a scTCR can be generated using methods known, See e.g., Soo Hoo, W. F. et al. PNAS (USA) 89, 4759 (1992 ); Wellerfing, C. and Plückthun, A., J. Mol. Biol. 242, 655 (1994 ); Kurucz, I. et al. PNAS (USA) 90 3830 (1993 ); International published PCT Nos. WO 96/13593 , WO 96/18105 , WO99/60120 , WO99/18129 , WO 03/020763 , WO2011/044186 ; and Schlueter, C. J.
  • a scTCR contains an introduced non-native disulfide interchain bond to facilitate the association of the TCR chains (see e.g. International published PCT No. WO 03/020763 ).
  • a scTCR is a non-disulfide linked truncated TCR in which heterologous leucine zippers fused to the C-termini thereof facilitate chain association (see e.g. International published PCT No. WO99/60120 ).
  • a scTCR contain a TCR ⁇ variable domain covalently linked to a TCR ⁇ variable domain via a peptide linker (see e.g., International published PCT No. WO99/18129 ).
  • a scTCR contains a first segment constituted by an amino acid sequence corresponding to a TCR ⁇ chain variable region, a second segment constituted by an amino acid sequence corresponding to a TCR ⁇ chain variable region sequence fused to the N terminus of an amino acid sequence corresponding to a TCR ⁇ chain constant domain extracellular sequence, and a linker sequence linking the C terminus of the first segment to the N terminus of the second segment.
  • a scTCR contains a first segment constituted by an ⁇ chain variable region sequence fused to the N terminus of an ⁇ chain extracellular constant domain sequence, and a second segment constituted by a ⁇ chain variable region sequence fused to the N terminus of a sequence ⁇ chain extracellular constant and transmembrane sequence, and, optionally, a linker sequence linking the C terminus of the first segment to the N terminus of the second segment.
  • a scTCR contains a first segment constituted by a TCR ⁇ chain variable region sequence fused to the N terminus of a ⁇ chain extracellular constant domain sequence, and a second segment constituted by an ⁇ chain variable region sequence fused to the N terminus of a sequence ⁇ chain extracellular constant and transmembrane sequence, and, optionally, a linker sequence linking the C terminus of the first segment to the N terminus of the second segment.
  • the linker of a scTCRs that links the first and second TCR segments can be any linker capable of forming a single polypeptide strand, while retaining TCR binding specificity.
  • the linker sequence may, for example, have the formula - P-AA-P- wherein P is proline and AA represents an amino acid sequence wherein the amino acids are glycine and serine.
  • the first and second segments are paired so that the variable region sequences thereof are orientated for such binding.
  • the linker has a sufficient length to span the distance between the C terminus of the first segment and the N terminus of the second segment, or vice versa, but is not too long to block or reduces bonding of the scTCR to the target ligand.
  • the linker can contain from or from about 10 to 45 amino acids, such as 10 to 30 amino acids or 26 to 41 amino acids residues, for example 29, 30, 31 or 32 amino acids.
  • the linker has the formula -PGGG-(SGGGG)5-P- wherein P is proline, G is glycine and S is serine (SEQ ID NO:32).
  • the linker has the sequence GSADDAKKDAAKKDGKS (SEQ ID NO:33).
  • the scTCR contains a covalent disulfide bond linking a residue of the immunoglobulin region of the constant domain of the ⁇ chain to a residue of the immunoglobulin region of the constant domain of the ⁇ chain.
  • the interchain disulfide bond in a native TCR is not present.
  • one or more cysteines can be incorporated into the constant region extracellular sequences of the first and second segments of the scTCR polypeptide. In some cases, both a native and a non-native disulfide bond may be desirable.
  • the native disulfide bonds are not present.
  • the one or more of the native cysteines forming a native interchain disulfide bonds are substituted to another residue, such as to a serine or alanine.
  • an introduced disulfide bond can be formed by mutating non-cysteine residues on the first and second segments to cysteine. Exemplary non-native disulfide bonds of a TCR are described in published International PCT No. WO2006/000830 .
  • the TCR or antigen-binding fragment thereof exhibits an affinity with an equilibrium binding constant for a target antigen of between or between about 10-5 and 10-12 M and all individual values and ranges therein.
  • the target antigen is an MHC-peptide complex or ligand.
  • nucleic acid or nucleic acids encoding a TCR can be amplified by PCR, cloning or other suitable means and cloned into a suitable expression vector or vectors.
  • the expression vector can be any suitable recombinant expression vector, and can be used to transform or transfect any suitable host. Suitable vectors include those designed for propagation and expansion or for expression or both, such as plasmids and viruses.
  • the vector can a vector of the pUC series (Fermentas Life Sciences), the pBluescript series (Stratagene, LaJolla, Calif.), the pET series (Novagen, Madison, Wis.), the pGEX series (Pharmacia Biotech, Uppsala, Sweden), or the pEX series (Clontech, Palo Alto, Calif.).
  • bacteriophage vectors such as ⁇ G10, ⁇ GT11, ⁇ ZapII (Stratagene), ⁇ EMBL4, and ⁇ NM1149, also can be used.
  • plant expression vectors can be used and include pBI01, pBI101.2, pBI101.3, pBI121 and pBIN19 (Clontech).
  • animal expression vectors include pEUK-Cl, pMAM and pMAMneo (Clontech).
  • a viral vector is used, such as a retroviral vector.
  • the recombinant expression vectors can be prepared using standard recombinant DNA techniques.
  • vectors can contain regulatory sequences, such as transcription and translation initiation and termination codons, which are specific to the type of host (e.g., bacterium, fungus, plant, or animal) into which the vector is to be introduced, as appropriate and taking into consideration whether the vector is DNA- or RNA-based.
  • the vector can contain a nonnative promoter operably linked to the nucleotide sequence encoding the TCR or antigen-binding portion (or other MHC-peptide binding molecule).
  • the promoter can be a non-viral promoter or a viral promoter, such as a cytomegalovirus (CMV) promoter, an SV40 promoter, an RSV promoter, and a promoter found in the long-terminal repeat of the murine stem cell virus.
  • CMV cytomegalovirus
  • SV40 SV40 promoter
  • RSV RSV promoter
  • promoter found in the long-terminal repeat of the murine stem cell virus a promoter found in the long-terminal repeat of the murine stem cell virus.
  • Other known promoters also are contemplated.
  • the ⁇ and ⁇ chains are PCR amplified from total cDNA isolated from a T cell clone expressing the TCR of interest and cloned into an expression vector.
  • the ⁇ and ⁇ chains are cloned into the same vector.
  • the ⁇ and ⁇ chains are cloned into different vectors.
  • the generated ⁇ and ⁇ chains are incorporated into a retroviral, e.g. lentiviral, vector.
  • an output cell composition is a cell composition that results from some or all of the steps for genetically engineering cells.
  • the output cell composition results from a process of genetically engineering cells of an input cell composition.
  • process contains one or more steps for activating, transducing or transfecting, expanding, and/or harvesting cells, such as cells that were obtained from an input cell composition.
  • the output cell composition contains cells that have been genetically engineered.
  • the cells of the output cell composition have undergone all of the steps for a process of genetic engineering.
  • the output cell composition contains cells that include one or more nucleic acids introduced via genetic engineering, and thereby express recombinant or genetically engineered products of such nucleic acids.
  • the nucleic acids are heterologous, i.e., normally not present in a cell or sample obtained from the cell, such as one obtained from another organism or cell, which for example, is not ordinarily found in the cell being engineered and/or an organism from which such cell is derived.
  • the nucleic acids are not naturally occurring, such as a nucleic acid not found in nature, including one comprising chimeric combinations of nucleic acids encoding various domains from multiple different cell types.
  • the output cell composition contains cells that have been genetically engineered.
  • the output cell composition contains engineered T cells.
  • the engineered T cells include engineered CD4+ T cells and engineered CD8+ T cells.
  • the output cell composition contains or includes at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70% , at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or 100% or about 100% engineered T cells.
  • the engineered cells express a recombinant receptor.
  • the output cell composition contains or includes at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70% , at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or 100% or about 100% T cells that express a recombinant receptor.
  • the recombinant receptor is a TCR or a CAR.
  • the recombinant receptor is a CAR.
  • the output cell composition has a ratio of engineered CD4+ T cells to CD8+ T cells of between 5:1 to 0.2:1, between 4:1 to 0.25:1, between 3:1 to 0.33:1, between 2:1 to 0.5:1, between 1.5:1 to 0.66:1, or between 1.25:1 to 0.8:1.
  • the output cell composition has a ratio of engineered CD4+ T cells to CD8+ T cells of between 2: 1 to 0.5:1.
  • the output cell composition has a ratio of engineered CD4+ T cells to CD8+ T cells of or of about 2.0:1, 1.9:1, 1.8:1, 1.7:1, 1.6:1, 1.5:1, 1.4:1, 1.3:1, 1.2:1, 1.1:1, 1.0:1, 0.9:1, 0.8:1, 0.7:1, 0.6:1, or 0.5:1.
  • the output cell composition has a ratio of engineered CD4+ T cells to CD8+ T cells of or of about 1:1.
  • the engineered T cells express a recombinant receptor.
  • the output cell composition has a ratio of recombinant receptor expressing CD4+ T cells to recombinant receptor expressing CD8+ T cells of or of about 2.0:1, 1.9:1, 1.8:1, 1.7:1, 1.6:1, 1.5:1, 1.4:1, 1.3:1, 1.2:1, 1.1:1, 1.0:1, 0.9:1, 0.8:1, 0.7:1, 0.6:1, or 0.5:1.
  • the output cell composition has a ratio of recombinant receptor expressing CD4+ T cells to recombinant receptor expressing CD8+ T cells of or of about 1:1.
  • the recombinant receptor is a TCR or a CAR.
  • the recombinant receptor is a CAR.
  • the methods result in an output cell composition having a ratio of engineered CD4+ T cells to engineered CD8+ T cells, a ratio of CD4+ T cells expressing a recombinant receptor to CD8+ T cells expressing a recombinant receptor, and/or a ratio of a ratio of CD4+ T cells expressing a CAR to CD8+ T cells expressing a CAR that is within a certain tolerated difference or range of error of such a defined, desired, or fixed ratio, and/or results in such a ratio a certain percentage of the time that the method is performed, such as at least at or about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or more than 95% of the time.
  • the tolerated difference is within about 1%, about 2%, about 3%, about 4% about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 60%, about 70%, or about 75% of the ratio.
  • the ratio is within 10%, 20%, or 30%, of the desired ratio and/or is within that ratio at least 70%, 80%, or 90% of the time the methods are performed. In certain instances, the ratio is within 50% of a 1:1 ratio at least 90% of the time the methods are performed.
  • the tolerated difference and/or the defined ratio of engineered, recombinant receptor expressing, and/or CAR expressing CD4+ to CD8+ T cells is or has been determined by administering different cell types, such as administering CD4 + and CD8 + T cells, to one or more subjects at a plurality of test ratios or numbers, and assessing one or more parameters.
  • the determination of the defined or fixed ratio or tolerated difference includes assessing one or more outcomes following administration to the subject.
  • the outcomes include those selected from among amelioration of a disease symptom and outcomes indicating safety and/or low or absence of toxicity.
  • an output cell composition produced by the methods disclosed herein has a ratio of engineered CD4+ T cells to engineered CD8+ T cells of between 2:1 and 0.5:1. In certain instances, the output composition contains a ratio of engineered CD4+ T cells to engineered CD8+ T cells of or of about 1:1. In some instances, the output cell composition was produced from an input cell composition described herein, e.g., an input cell composition described in Section I-A, and has a ratio of engineered CD4+ T cells to engineered CD8+ T cells of between 2:1 and 0.5:1.
  • the output cell composition produced from an input cell composition has a ratio of engineered CD4+ T cells to engineered CD8+ T cells of 1:1 with a tolerated different of 50%, 25%, 10%, or less.
  • the engineered T cells express a recombinant receptor.
  • the engineered T cells express a CAR.
  • compositions or formulations containing cells prepared according to the methods of incubating (e.g., stimulating) described herein.
  • the compositions and methods described herein can be used to obtain an output cell composition of cells with a defined ratio of recombinant receptor expressing CD4+ to CD8+ T cells, such as for use as a therapeutic cell composition.
  • an output cell composition produced by any of the methods described herein.
  • the cells produced using any of the methods described herein are disclosed as compositions, including pharmaceutical compositions and formulations, such as unit dose form compositions including the number of cells for administration in a given dose or fraction thereof.
  • the cells of the output cell composition are genetically engineered with a recombinant receptor (e.g., CAR-T cells).
  • the pharmaceutical compositions and formulations generally include one or more optional pharmaceutically acceptable carrier or excipient.
  • the composition includes at least one additional therapeutic agent.
  • a composition of cells e.g., an output cell composition
  • the cell composition is a pharmaceutical composition or formulation.
  • Such compositions can be used in accord with the disclosed methods, for example, to assess their release for use in the prevention or treatment of diseases, conditions, and disorders, or in detection, diagnostic, and prognostic methods.
  • pharmaceutical formulation refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • methods disclosed herein may be used to compare surface glycan expression of cell compositions composed of the same engineered cells, but with different pharmaceutical formulations.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative. In particular instances, methods disclosed herein may be used to compare surface glycan expression of cell compositions composed of the same engineered cells, but with different pharmaceutically acceptable carriers.
  • the T cell therapy such as engineered T cells (e.g., CAR T cells) are formulated with a pharmaceutically acceptable carrier.
  • the choice of carrier is determined in part by the particular cell and/or by the method of administration. Accordingly, there are a variety of suitable formulations.
  • the pharmaceutical composition can contain preservatives. Suitable preservatives may include, for example, methylparaben, propylparaben, sodium benzoate, and benzalkonium chloride. In some aspects, a mixture of two or more preservatives is used. The preservative or mixtures thereof are typically present in an amount of about 0.0001% to about 2% by weight of the total composition.
  • Carriers are described, e.g., by Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980 ).
  • Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers
  • Buffering agents in some aspects are included in the compositions. Suitable buffering agents include, for example, citric acid, sodium citrate, phosphoric acid, potassium phosphate, and various other acids and salts. In some aspects, a mixture of two or more buffering agents is used. The buffering agent or mixtures thereof are typically present in an amount of about 0.001% to about 4% by weight of the total composition. Methods for preparing administrable pharmaceutical compositions are known. Exemplary methods are described in more detail in, for example, Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins; 21st ed. (May 1, 2005 ).
  • the formulations can include aqueous solutions.
  • the formulation or composition may also contain more than one active ingredient useful for the particular indication, disease, or condition being prevented or treated with the cells, including one or more active ingredients where the activities are complementary to the cells and/or the respective activities do not adversely affect one another.
  • active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
  • the pharmaceutical composition further includes other pharmaceutically active agents or drugs, such as chemotherapeutic agents, e.g., asparaginase, busulfan, carboplatin, cisplatin, daunorubicin, doxorubicin, fluorouracil, gemcitabine, hydroxyurea, methotrexate, paclitaxel, rituximab, vinblastine, vincristine, etc.
  • chemotherapeutic agents e.g., asparaginase, busulfan, carboplatin, cisplatin, daunorubicin, doxorubicin, fluorouracil, gemcitabine, hydroxyurea, methotrexate, paclitaxel, rituximab, vinblastine, vincristine, etc.
  • the pharmaceutical composition in some instances, contain cells, e.g., cells of the output cell composition, in amounts effective to treat or prevent the disease or condition, such as a therapeutically effective or prophylactically effective amount.
  • Therapeutic or prophylactic efficacy in some instances is monitored by periodic assessment of treated subjects. For repeated administrations over several days or longer, depending on the condition, the treatment is repeated until a desired suppression of disease symptoms occurs.
  • other dosage regimens may be useful and can be determined.
  • the desired dosage can be delivered by a single bolus administration of the composition, by multiple bolus administrations of the composition, or by continuous infusion administration of the composition.
  • the cells may be formulated for administration using standard administration techniques, formulations, and/or devices.
  • formulations and devices such as syringes and vials, for storage and administration of the compositions.
  • administration can be autologous or heterologous.
  • immunoresponsive cells or progenitors can be obtained from one subject, and administered to the same subject or a different, compatible subject.
  • Peripheral blood derived immunoresponsive cells or their progeny e.g., in vivo, ex vivo or in vitro derived
  • can be administered via localized injection including catheter administration, systemic injection, localized injection, intravenous injection, or parenteral administration.
  • a therapeutic composition e.g., a pharmaceutical composition containing a genetically modified immunoresponsive cell
  • it will generally be formulated in a unit dosage injectable form (solution, suspension, emulsion).
  • Formulations include those for oral, intravenous, intraperitoneal, subcutaneous, pulmonary, transdermal, intramuscular, intranasal, buccal, sublingual, or suppository administration.
  • the agent or cell populations are administered parenterally.
  • parenteral includes intravenous, intramuscular, subcutaneous, rectal, vaginal, and intraperitoneal administration.
  • the agent or cell populations are administered to a subject using peripheral systemic delivery by intravenous, intraperitoneal, or subcutaneous injection.
  • compositions in some instances are disclosed as sterile liquid preparations, e.g., isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous compositions, which may in some aspects be buffered to a selected pH.
  • sterile liquid preparations e.g., isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous compositions, which may in some aspects be buffered to a selected pH.
  • Liquid preparations are normally easier to prepare than gels, other viscous compositions, and solid compositions. Additionally, liquid compositions are somewhat more convenient to administer, especially by injection. Viscous compositions, on the other hand, can be formulated within the appropriate viscosity range to provide longer contact periods with specific tissues.
  • Liquid or viscous compositions can comprise carriers, which can be a solvent or dispersing medium containing, for example, water, saline, phosphate buffered saline, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol) and suitable mixtures thereof.
  • carriers can be a solvent or dispersing medium containing, for example, water, saline, phosphate buffered saline, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol) and suitable mixtures thereof.
  • Sterile injectable solutions can be prepared by incorporating the cells in a solvent, such as in admixture with a suitable carrier, diluent, or excipient such as sterile water, physiological saline, glucose, dextrose, or the like.
  • a suitable carrier such as a suitable carrier, diluent, or excipient
  • the compositions can also be lyophilized.
  • the compositions can contain auxiliary substances such as wetting, dispersing, or emulsifying agents (e.g., methylcellulose), pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired. Standard texts may in some aspects be consulted to prepare suitable preparations.
  • compositions including antimicrobial preservatives, antioxidants, chelating agents, and buffers, can be added.
  • antimicrobial preservatives for example, parabens, chlorobutanol, phenol, sorbic acid, and the like.
  • Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • the formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
  • the appropriate dosage may depend on the type of disease to be treated, the type of agent or agents, the type of cells or recombinant receptors, the severity and course of the disease, whether the agent or cells are administered for preventive or therapeutic purposes, previous therapy, the subject's clinical history and response to the agent or the cells, and the discretion of the attending physician.
  • the compositions are in some instances suitably administered to the subject at one time or over a series of treatments.
  • the disclosed methods and uses include methods and uses for adoptive cell therapy.
  • the methods include administration of the engineered cells or a composition containing the cells, such as cells from an output composition as described, to a subject, tissue, or cell, such as one having, at risk for, or suspected of having the disease, condition or disorder.
  • the cells, populations, and compositions are administered to a subject having the particular disease or condition to be treated, e.g., via adoptive cell therapy, such as adoptive T cell therapy.
  • the cells or compositions are administered to the subject, such as a subject having or at risk for the disease or condition, ameliorate one or more symptom of the disease or condition, such as by lessening tumor burden in a cancer expressing an antigen recognized by an engineered T cell.
  • the disease or condition that is treated in some aspects can be any in which expression of an antigen is associated with, specific to, and/or expressed on a cell or tissue of a disease, disorder or condition and/or involved in the etiology of a disease, condition or disorder, e.g. causes, exacerbates or otherwise is involved in such disease, condition, or disorder.
  • exemplary diseases and conditions can include diseases or conditions associated with malignancy or transformation of cells (e.g. cancer), autoimmune or inflammatory disease, or an infectious disease, e.g. caused by a bacterial, viral or other pathogen.
  • Exemplary antigens which include antigens associated with various diseases and conditions that can be treated, are described above.
  • the immunomodulatory polypeptide and/or recombinant receptor e.g., the chimeric antigen receptor or TCR, specifically binds to an antigen associated with the disease or condition.
  • the subject has a disease, disorder or condition, optionally a cancer, a tumor, an autoimmune disease, disorder or condition, or an infectious disease.
  • the disease, disorder or condition includes tumors associated with various cancers.
  • the cancer can in some instances be any cancer located in the body of a subject, such as, but not limited to, cancers located at the head and neck, breast, liver, colon, ovary, prostate, pancreas, brain, cervix, bone, skin, eye, bladder, stomach, esophagus, peritoneum, or lung.
  • the anti-cancer agent can be used for the treatment of colon cancer, cervical cancer, cancer of the central nervous system, breast cancer, bladder cancer, anal carcinoma, head and neck cancer, ovarian cancer, endometrial cancer, small cell lung cancer, non-small cell lung carcinoma, neuroendocrine cancer, soft tissue carcinoma, penile cancer, prostate cancer, pancreatic cancer, gastric cancer, gall bladder cancer or espohageal cancer.
  • the cancer can be a cancer of the blood.
  • the disease, disorder or condition is a tumor, such as a solid tumor, lymphoma, leukemia, blood tumor, metastatic tumor, or other cancer or tumor type.
  • the disease, disorder or condition is selected from among cancers of the colon, lung, liver, breast, prostate, ovarian, skin, melanoma, bone, brain cancer, ovarian cancer, epithelial cancers, renal cell carcinoma, pancreatic adenocarcinoma, cervical carcinoma, colorectal cancer, glioblastoma, neuroblastoma, Ewing sarcoma, medulloblastoma, osteosarcoma, synovial sarcoma, and/or mesothelioma.
  • the diseases, conditions, and disorders are tumors, including solid tumors, hematologic malignancies, and melanomas, and including localized and metastatic tumors, infectious diseases, such as infection with a virus or other pathogen, e.g., HIV, HCV, HBV, CMV, HPV, and parasitic disease, and autoimmune and inflammatory diseases.
  • infectious diseases such as infection with a virus or other pathogen, e.g., HIV, HCV, HBV, CMV, HPV, and parasitic disease
  • autoimmune and inflammatory diseases e.g., a virus or other pathogen, e.g., HIV, HCV, HBV, CMV, HPV, and parasitic disease
  • the disease, disorder or condition is a tumor, cancer, malignancy, neoplasm, or other proliferative disease or disorder.
  • Such diseases include but are not limited to leukemia, lymphoma, e.g., acute myeloid (or myelogenous) leukemia (AML), chronic myeloid (or myelogenous) leukemia (CML), acute lymphocytic (or lymphoblastic) leukemia (ALL), chronic lymphocytic leukemia (CLL), hairy cell leukemia (HCL), small lymphocytic lymphoma (SLL), Mantle cell lymphoma (MCL), Marginal zone lymphoma, Burkitt lymphoma, Hodgkin lymphoma (HL), non-Hodgkin lymphoma (NHL), Anaplastic large cell lymphoma (ALCL), follicular lymphoma, refractory follicular lymphoma, diffuse large B-cell lymphoma (DLBCL) and multiple myeloma (MM), a B cell malignancy is selected from among acute lymphoblastic leukemia (ALL), adult ALL,
  • the disease or condition is an infectious disease or condition, such as, but not limited to, viral, retroviral, bacterial, and protozoal infections, immunodeficiency, Cytomegalovirus (CMV), Epstein-Barr virus (EBV), adenovirus, BK polyomavirus.
  • infectious disease or condition such as, but not limited to, viral, retroviral, bacterial, and protozoal infections, immunodeficiency, Cytomegalovirus (CMV), Epstein-Barr virus (EBV), adenovirus, BK polyomavirus.
  • the disease or condition is an autoimmune or inflammatory disease or condition, such as arthritis, e.g., rheumatoid arthritis (RA), Type I diabetes, systemic lupus erythematosus (SLE), inflammatory bowel disease, psoriasis, scleroderma, autoimmune thyroid disease, Grave's disease, Crohn's disease, multiple sclerosis, asthma, and/or a disease or condition associated with transplant.
  • arthritis e.g., rheumatoid arthritis (RA), Type I diabetes, systemic lupus erythematosus (SLE), inflammatory bowel disease, psoriasis, scleroderma, autoimmune thyroid disease, Grave's disease, Crohn's disease, multiple sclerosis, asthma, and/or a disease or condition associated with transplant.
  • RA rheumatoid arthritis
  • SLE systemic lupus erythematosus
  • inflammatory bowel disease e.
  • the antigen associated with the disease or disorder is selected from the group consisting of orphan tyrosine kinase receptor ROR1, B cell maturation antigen (BCMA), carbonic anhydrase 9 (CAIX), tEGFR, Her2/neu (receptor tyrosine kinase erbB2), L1-CAM, CD19, CD20, CD22, mesothelin, CEA, hepatitis B surface antigen, anti-folate receptor, CD23, CD24, CD30, CD33, CD38, CD44, EGFR, epithelial glycoprotein 2 (EPG-2), epithelial glycoprotein 40 (EPG-40), ephrine receptor A2 (EPHa2), Her2/neu (receptor tyrosine kinase erb-B2), Her3 (erb-B3), Her4 (erb-B4), erbB dimers, type III epidermal growth factor receptor mutation (EGFR vIII), fo
  • the antigen or ligand is a tumor antigen or cancer marker.
  • the antigen or ligand the antigen is or includes orphan tyrosine kinase receptor (ROR1), B cell maturation antigen (BCMA), carbonic anhydrase 9 (CA9, also known as CAIX or G250), Her2/neu (receptor tyrosine kinase erbB2), CD19, CD20, CD22, mesothelin (MSLN), carcinoembryonic antigen (CEA), and hepatitis B surface antigen, anti-folate receptor, CD23, CD24, CD30, CD33, CD38, CD44, chondroitin sulfate proteoglycan 4 (CSPG4), EGFR, epithelial glycoprotein 2 (EPG-2), epithelial glycoprotein 40 (EPG-40), ephrin receptor A2 (EPHa2), Her3 (erb-B3), Her4 (er
  • ROR1
  • the disease or condition is a B cell malignancy.
  • the B cell malignancy is a leukemia or a lymphoma.
  • the disease or condition is acute lymphoblastic leukemia (ALL), adult ALL, chronic lymphoblastic leukemia (CLL), non-Hodgkin lymphoma (NHL), or Diffuse Large B-Cell Lymphoma (DLBCL).
  • ALL acute lymphoblastic leukemia
  • CLL chronic lymphoblastic leukemia
  • NHL non-Hodgkin lymphoma
  • the disease or condition is an NHL, such as or including an NHL that is an aggressive NHL, diffuse large B cell lymphoma (DLBCL), NOS (de novo and transformed from indolent), primary mediastinal large B cell lymphoma (PMBCL), T cell/histocyte-rich large B cell lymphoma (TCHRBCL), Burkitt's lymphoma, mantle cell lymphoma (MCL), and/or follicular lymphoma (FL), optionally, follicular lymphoma Grade 3B (FL3B).
  • NHL such as or including an NHL that is an aggressive NHL, diffuse large B cell lymphoma (DLBCL), NOS (de novo and transformed from indolent), primary mediastinal large B cell lymphoma (PMBCL), T cell/histocyte-rich large B cell lymphoma (TCHRBCL), Burkitt's lymphoma, mantle cell lymphoma (MCL), and/or follicular lymphoma (
  • the recombinant receptor such as a CAR, specifically binds to an antigen associated with the disease or condition or expressed in cells of the environment of a lesion associated with the B cell malignancy.
  • Antigens targeted by the receptors in some instances include antigens associated with a B cell malignancy, such as any of a number of known B cell marker.
  • the antigen targeted by the receptor is CD20, CD19, CD22, ROR1, CD45, CD21, CD5, CD33, Igkappa, Iglambda, CD79a, CD79b or CD30.
  • the disease or condition is a myeloma, such as a multiple myeloma.
  • the recombinant receptor such as a CAR, specifically binds to an antigen associated with the disease or condition or expressed in cells of the environment of a lesion associated with the multiple myeloma.
  • Antigens targeted by the receptors in some instances include antigens associated with multiple myeloma, such as GPRC5D or BCMA.
  • the antigen is a pathogen-specific or pathogen-expressed antigen.
  • the antigen is a viral antigen (such as a viral antigen from HIV, HCV, HBV, etc.), bacterial antigens, and/or parasitic antigens.
  • the cell-based therapy is or comprises administration of cells, such as T cells, that target a molecule expressed on the surface of a lesion, such as a tumor or a cancer.
  • the immune cells express a T cell receptor (TCR) or other antigen-binding receptor.
  • the immune cells express a recombinant receptor, such as a transgenic TCR or a chimeric antigen receptor (CAR).
  • the cells are autologous to the subject.
  • the cells are allogeneic to the subject.
  • the cell therapy e.g., adoptive T cell therapy
  • the cell therapy is carried out by autologous transfer, in which the cells are isolated and/or otherwise prepared from the subject who is to receive the cell therapy, or from a sample derived from such a subject.
  • the cells are derived from a subject, e.g ., patient, in need of a treatment and the cells, following isolation and processing are administered to the same subject.
  • the cell therapy e.g ., adoptive T cell therapy
  • the cells are carried out by allogeneic transfer, in which the cells are isolated and/or otherwise prepared from a subject other than a subject who is to receive or who ultimately receives the cell therapy, e.g ., a first subject.
  • the cells then are administered to a different subject, e.g ., a second subject, of the same species.
  • the first and second subjects are genetically identical.
  • the first and second subjects are genetically similar.
  • the second subject expresses the same HLA class or supertype as the first subject.
  • the cells can be administered by any suitable means. Dosing and administration may depend in part on whether the administration is brief or chronic. Various dosing schedules include but are not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion.
  • the disclosed methods include one or more steps administering to a subject cells of the output cell composition, such as a composition of cells described in Section I.
  • the cells of the output cell composition include engineered CD4+ T cells and engineered CD8+ T cells.
  • the engineered CD4+ and CD8+ T cells express a T cell receptor (TCR) or other antigen-binding receptor.
  • the immune cells express a recombinant receptor, such as a transgenic TCR or a chimeric antigen receptor (CAR).
  • the cells of the output cell composition are autologous to the subject. In some instances, the cells are allogeneic to the subject.
  • the CD4+ T cells and CD8+ T cells of the output cell composition are administered to the subject in the same composition, dose, or mixture.
  • the recombinant receptor expressing CD4+ T cells and recombinant receptor expressing CD8+ T cells e.g., CAR+CD4+ and CAR+CD8+ are administered to the subject in the same composition, dose, or mixture.
  • the cells are administered to the subject at a range of about one million to about 100 billion cells and/or that amount of cells per kilogram of body weight, such as, e.g., 1 million to about 50 billion cells (e.g., about 5 million cells, about 25 million cells, about 500 million cells, about 1 billion cells, about 5 billion cells, about 20 billion cells, about 30 billion cells, about 40 billion cells, or a range defined by any two of the foregoing values), such as about 10 million to about 100 billion cells (e.g., about 20 million cells, about 30 million cells, about 40 million cells, about 60 million cells, about 70 million cells, about 80 million cells, about 90 million cells, about 10 billion cells, about 25 billion cells, about 50 billion cells, about 75 billion cells, about 90 billion cells, or a range defined by any two of the foregoing values), and in some cases about 100 million cells to about 50 billion cells (e.g., about 120 million cells, about
  • the dose includes fewer than about 1 ⁇ 10 8 total recombinant receptor (e.g., CAR)-expressing cells, T cells, or peripheral blood mononuclear cells (PBMCs), e.g., in the range of about 1 ⁇ 10 6 to 1 ⁇ 10 8 such cells, such as 2 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , or 1 ⁇ 10 8 or total such cells, or the range between any two of the foregoing values.
  • CAR total recombinant receptor
  • PBMCs peripheral blood mononuclear cells
  • the dose of genetically engineered cells comprises from or from about 1 ⁇ 10 5 to 5 ⁇ 10 8 total CAR-expressing T cells, 1 ⁇ 10 5 to 2.5 ⁇ 10 8 total CAR-expressing T cells, 1 ⁇ 10 5 to 1 ⁇ 10 8 total CAR-expressing T cells, 1 ⁇ 10 5 to 5 ⁇ 10 7 total CAR-expressing T cells, 1 ⁇ 10 5 to 2.5 ⁇ 10 7 total CAR-expressing T cells, 1 ⁇ 10 5 to 1 ⁇ 10 7 total CAR-expressing T cells, 1 ⁇ 10 5 to 5 ⁇ 10 6 total CAR-expressing T cells, 1 ⁇ 10 5 to 2.5 ⁇ 10 6 total CAR-expressing T cells, 1 ⁇ 10 5 to 1 ⁇ 10 6 total CAR-expressing T cells, 1 ⁇ 10 6 to 5 ⁇ 10 8 total CAR-expressing T cells, 1 ⁇ 10 6 to 2.5 ⁇ 10 8 total CAR-expressing T cells, 1 ⁇ 10 6 to 1 ⁇ 10 8 total CAR-expressing T cells, 1 ⁇ 10 6 to 5 ⁇ 10 8 total C
  • the dose of genetically engineered cells comprises at least or at least about 1 ⁇ 10 5 CAR-expressing cells, at least or at least about 2.5 ⁇ 10 5 CAR-expressing cells, at least or at least about 5 ⁇ 10 5 CAR-expressing cells, at least or at least about 1 ⁇ 10 6 CAR-expressing cells, at least or at least about 2.5 ⁇ 10 6 CAR-expressing cells, at least or at least about 5 ⁇ 10 6 CAR-expressing cells, at least or at least about 1 ⁇ 10 7 CAR-expressing cells, at least or at least about 2.5 ⁇ 10 7 CAR-expressing cells, at least or at least about 5 ⁇ 10 7 CAR-expressing cells, at least or at least about 1 ⁇ 10 8 CAR-expressing cells, at least or at least about 2.5 ⁇ 10 8 CAR-expressing cells, or at least or at least about 5 ⁇ 10 8 CAR-expressing cells.
  • the cell therapy comprises administration of a dose comprising a number of cell from or from about 1 ⁇ 10 5 to 5 ⁇ 10 8 total recombinant receptor-expressing cells, total T cells, or total peripheral blood mononuclear cells (PBMCs), from or from about 5 ⁇ 10 5 to 1 ⁇ 10 7 total recombinant receptor-expressing cells, total T cells, or total peripheral blood mononuclear cells (PBMCs) or from or from about 1 ⁇ 10 6 to 1 ⁇ 10 7 total recombinant receptor-expressing cells, total T cells, or total peripheral blood mononuclear cells (PBMCs), each inclusive.
  • PBMCs peripheral blood mononuclear cells
  • the cell therapy comprises administration of a dose of cells comprising a number of cells at least or at least about 1 ⁇ 10 5 total recombinant receptor-expressing cells, total T cells, or total peripheral blood mononuclear cells (PBMCs), such at least or at least 1 ⁇ 10 6 , at least or at least about 1 ⁇ 10 7 , at least or at least about 1 ⁇ 10 8 of such cells.
  • the number is with reference to the total number of CD3+ or CD8+, in some cases also recombinant receptor-expressing (e.g. CAR+) cells.
  • the cell therapy comprises administration of a dose comprising a number of cell from or from about 1 ⁇ 10 5 to 5 ⁇ 10 8 CD3+ or CD8+ total T cells or CD3+ or CD8+ recombinant receptor-expressing cells, from or from about 5 ⁇ 10 5 to 1 ⁇ 10 7 CD3+ or CD8+ total T cells or CD3+ or CD8+ recombinant receptor-expressing cells, or from or from about 1 ⁇ 10 6 to 1 ⁇ 10 7 CD3+ or CD8+ total T cells or CD3+ or CD8+recombinant receptor-expressing cells, each inclusive.
  • the cell therapy comprises administration of a dose comprising a number of cell from or from about 1 ⁇ 10 5 to 5 ⁇ 10 8 total CD3+/CAR+ or CD8+/CAR+ cells, from or from about 5 ⁇ 10 5 to 1 ⁇ 10 7 total CD3+/CAR+ or CD8+/CAR+ cells, or from or from about 1 ⁇ 10 6 to 1 ⁇ 10 7 total CD3+/CAR+ or CD8+/CAR+ cells, each inclusive.
  • the CD8+ T cells of the dose includes between about 1 ⁇ 10 6 and 5 ⁇ 10 8 total recombinant receptor (e.g., CAR)-expressing CD8+cells, e.g., in the range of about 5 ⁇ 10 6 to 1 ⁇ 10 8 such cells, such cells 1 ⁇ 10 7 , 2.5 ⁇ 10 7 , 5 ⁇ 10 7 , 7.5 ⁇ 10 7 , 1 ⁇ 10 8 , or 5 ⁇ 10 8 total such cells, or the range between any two of the foregoing values.
  • CAR total recombinant receptor
  • the patient is administered multiple doses, and each of the doses or the total dose can be within any of the foregoing values.
  • the dose of cells comprises the administration of from or from about 1 ⁇ 10 7 to 0.75 ⁇ 10 8 total recombinant receptor-expressing CD8+ T cells, 1 ⁇ 10 7 to 2.5 ⁇ 10 7 total recombinant receptor-expressing CD8+ T cells, from or from about 1 ⁇ 10 7 to 0.75 ⁇ 10 8 total recombinant receptor-expressing CD8+ T cells, each inclusive.
  • the dose of cells comprises the administration of or about 1 ⁇ 10', 2.5 ⁇ 10 7 , 5 ⁇ 10 7 7.5 ⁇ 10 7 , 1 ⁇ 10 8 , or 5 ⁇ 10 8 total recombinant receptor-expressing CD8+ T cells.
  • the dose of cells e.g., recombinant receptor-expressing T cells
  • the pharmaceutical compositions and formulations are disclosed as unit dose form compositions including the number of cells for administration in a given dose or fraction thereof.
  • the disclosed methods produce cells in a predictable timeline to dosing as compared to other methods of incubating (e.g., stimulating) cells.
  • the dose of cells for administration is determined based on the number of na ⁇ ve-like cells in the input cell composition.
  • the size of the dose is determined by the burden of the disease or condition in the subject. For example, in some aspects, the number of cells administered in the dose is determined based on the tumor burden that is present in the subject immediately prior to administration of the initiation of the dose of cells. In some instances, the size of the first and/or subsequent dose is inversely correlated with disease burden. In some aspects, as in the context of a large disease burden, the subject is administered a low number of cells. In other instances, as in the context of a lower disease burden, the subject is administered a larger number of cells.
  • the biological activity of the engineered cell populations in some instances is measured, e.g., by any of a number of known methods.
  • Parameters to assess include specific binding of an engineered or natural T cell or other immune cell to antigen, in vivo, e.g., by imaging, or ex vivo, e.g., by ELISA or flow cytometry.
  • the ability of the engineered cells to destroy target cells can be measured using any suitable method, such as cytotoxicity assays described in, for example, Kochenderfer et al., J. Immunotherapy, 32(7): 689-702 (2009 ), and Herman et al. J. Immunological Methods, 285(1): 25-40 (2004 ).
  • the biological activity of the cells is measured by assaying expression and/or secretion of one or more cytokines, such as CD 107a, IFN ⁇ , IL-2, and TNF. In some aspects the biological activity is measured by assessing clinical outcome, such as reduction in tumor burden or load.
  • cytokines such as CD 107a, IFN ⁇ , IL-2, and TNF.
  • the cells can be administered by any suitable means.
  • the cells are administered in a dosing regimen to achieve a therapeutic effect, such as a reduction in tumor burden. Dosing and administration may depend in part on the schedule of administration of the immunomodulatory compound, which can be administered prior to, subsequent to and/or simultaneously with initiation of administration of the T cell therapy.
  • Various dosing schedules of the T cell therapy include but are not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion.
  • the engineered T cells express a recombinant receptor.
  • the engineered T cells express a CAR.
  • the ratio of the CD4+ T cells to CD8+ T cells that are administered to the subject in the same composition, dose, or mixture is between 5:1 to 0.2:1, between 4:1 to 0.25:1, between 3:1 to 0.33:1, between 2:1 to 0.5:1, between 1.5:1 to 0.66:1, or between 1.25:1 to 0.8:1.
  • the ratio of CD4+ T cells to CD8+ T cells administered to the subject in the same composition, dose, or mixture is or is about 2.0:1, 1.9:1, 1.8:1, 1.7:1, 1.6:1, 1.5:1, 1.4:1, 1.3:1, 1.2:1, 1.1:1, 1.0:1, 0.9:1, 0.8:1, 0.7:1, 0.6:1, or 0.5:1.
  • the ratio of recombinant receptor expressing CD4+ T cells to recombinant receptor expressing CD8+ T cells that are administered to the subject in the same composition, dose, or mixture is between 5:1 and 0.2:1, between 4:1 and 0.25:1, between 3:1 and 0.33:1, between 2:1 and 0.5:1, between 1.5:1 and 0.66:1, or between 1.25:1 and 0.8:1.
  • the ratio of recombinant receptor expressing CD4+ T cells to recombinant receptor expressing CD8+ T cells that are administered to the subject in the same composition, dose, or mixture is or is about 2.0:1, 1.9:1, 1.8:1, 1.7:1, 1.6:1, 1.5:1, 1.4:1, 1.3:1, 1.2:1, 1.1:1, 1.0:1, 0.9:1, 0.8:1, 0.7:1, 0.6:1, or 0.5:1.
  • ratio of the administered recombinant receptor expressing CD4+ T cells to recombinant receptor expressing CD8+ T cells is or is about 1:1.
  • the recombinant receptor is a TCR or a CAR.
  • the recombinant receptor is a CAR.
  • the ratio of engineered CD4+ T cells to engineered CD8+ T cells of the dose, composition, or mixture that is administered to the subject is within a certain tolerated difference or range of error of such a defined, desired, or fixed ratio.
  • the tolerated difference is within of or of about 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50%, of the target, defined, preferred, and/or fixed ratio.
  • a composition of cells produced by the methods disclosed herein having a ratio of engineered CD4+ T cells to engineered CD8+ T cells of between 2:1 and 0.5:1 is administered to a subject in a single composition, dose, or mixture.
  • the composition contains a ratio of engineered CD4+ T cells to engineered CD8+ T cells of or of about 1:1.
  • a cell composition produced from an input cell composition e.g., an input cell composition described in Section I-A1
  • the cell composition produced from an input cell composition has a ratio of engineered CD4+ T cells to engineered CD8+ T cells of 1:1 with a tolerated difference of 50%, 25%, 10%, or less.
  • the cells are administered as part of a further combination treatment, such as simultaneously with or sequentially with, in any order, another therapeutic intervention, such as an antibody or engineered cell or receptor or agent, such as a cytotoxic or therapeutic agent.
  • another therapeutic intervention such as an antibody or engineered cell or receptor or agent, such as a cytotoxic or therapeutic agent.
  • an anti-cancer agent or immunomodulatory agent can be used in combination therapy with adoptive cell therapy with engineered cell expressing a recombinant receptor, e.g. a CAR.
  • the cells are co-administered with another therapy sufficiently close in time such that the cell populations enhance the effect of the P one or more additional therapeutic agents, or vice versa.
  • the cells are administered prior to the one or more additional therapeutic agents.
  • the cells are administered after the one or more additional therapeutic agents.
  • the one or more additional therapeutic agents include a cytokine, such as IL-2, for example, to enhance persistence.
  • the methods comprise administration of a chemotherapeutic agent.
  • the one or more additional therapeutic agents include one or more lymphodepleting therapies, such as prior to or simultaneous with initiation of administration of the engineered cells.
  • the lymphodepleting therapy comprises administration of a phosphamide, such as cyclophosphamide.
  • the lymphodepleting therapy can include administration of fludarabine.
  • fludarabine is excluded in the lymphodepleting therapy.
  • a lymphodepleting therapy is not administered.
  • the methods include administering a preconditioning agent, such as a lymphodepleting or chemotherapeutic agent, such as cyclophosphamide, fludarabine, or combinations thereof, to a subject prior to the initiation of the cell therapy.
  • a preconditioning agent such as a lymphodepleting or chemotherapeutic agent, such as cyclophosphamide, fludarabine, or combinations thereof.
  • the subject may be administered a preconditioning agent at least 2 days prior, such as at least 3, 4, 5, 6, or 7 days prior, to the initiation of the cell therapy.
  • the subject is administered a preconditioning agent no more than 7 days prior, such as no more than 6, 5, 4, 3, or 2 days prior, to the initiation of the cell therapy.
  • the subject is preconditioned with cyclophosphamide at a dose between or between about 20 mg/kg and 100 mg/kg, such as between or between about 40 mg/kg and 80 mg/kg. In some aspects, the subject is preconditioned with or with about 60 mg/kg of cyclophosphamide.
  • the cyclophosphamide can be administered in a single dose or can be administered in a plurality of doses, such as given daily, every other day or every three days. In some instances, the cyclophosphamide is administered once daily for one or two days.
  • the subject is administered cyclophosphamide at a dose between or between about 100 mg/m 2 and 500 mg/m 2 , such as between or between about 200 mg/m 2 and 400 mg/m 2 , or 250 mg/m 2 and 350 mg/m 2 , inclusive. In some instances, the subject is administered about 300 mg/m 2 of cyclophosphamide. In some instances, the cyclophosphamide can be administered in a single dose or can be administered in a plurality of doses, such as given daily, every other day or every three days.
  • cyclophosphamide is administered daily, such as for 1-5 days, for example, for 3 to 5 days. In some instances, the subject is administered about 300 mg/m 2 of cyclophosphamide, daily for 3 days, prior to initiation of the cell therapy.
  • the subject is administered fludarabine at a dose between or between about 1 mg/m 2 and 100 mg/m 2 , such as between or between about 10 mg/m 2 and 75 mg/m 2 , 15 mg/m 2 and 50 mg/m 2 , 20 mg/m 2 and 40 mg/m 2 , or 24 mg/m 2 and 35 mg/m 2 , inclusive.
  • the subject is administered about 30 mg/m 2 of fludarabine.
  • the fludarabine can be administered in a single dose or can be administered in a plurality of doses, such as given daily, every other day or every three days.
  • fludarabine is administered daily, such as for 1-5 days, for example, for 3 to 5 days.
  • the subject is administered about 30 mg/m 2 of fludarabine, daily for 3 days, prior to initiation of the cell therapy.
  • the lymphodepleting agent comprises a combination of agents, such as a combination of cyclophosphamide and fludarabine.
  • the combination of agents may include cyclophosphamide at any dose or administration schedule, such as those described above, and fludarabine at any dose or administration schedule, such as those described above.
  • the subject is administered 60 mg/kg ( ⁇ 2 g/m 2 ) of cyclophosphamide and 3 to 5 doses of 25 mg/m 2 fludarabine prior to the first or subsequent dose.
  • kits and devices for the administration of the cells to subjects in according to the disclosed methods for adoptive cell therapy, and for storage and administration of the cells and compositions, such as the input compositions or output compositions as described.
  • the articles of manufacture include one or more containers, typically a plurality of containers, packaging material, and a label or package insert on or associated with the container or containers and/or packaging, generally including instructions for administration of the cells to a subject.
  • the containers contain the cells to be administered, e.g., one or more unit doses thereof.
  • the article of manufacture typically includes a plurality of containers, each containing a single unit dose of the cells.
  • the unit dose may be an amount or number of the cells to be administered to the subject in the first dose or twice the number (or more) the cells to be administered in the first or any one or more consecutive dose(s). It may be the lowest dose or lowest possible dose of the cells that would be administered to the subject in connection with the administration method.
  • the unit dose is the minimum number of cells or number of cells or the minimum number of reference units or the target reference units or reference units within a target range that would be administered in a single dose to any subject having a particular disease or condition or any subject, according to the methods herein.
  • Suitable containers include, for example, bottles, vials, syringes, and flexible bags, such as infusion bags.
  • the containers are bags, e.g., flexible bags, such as those suitable for infusion of cells to subjects, e.g., flexible plastic or PVC bags, and/or IV solution bags.
  • the bags in some instances are sealable and/or able to be sterilized, so as to provide sterile solution and delivery of the cells and compositions.
  • the containers e.g., bags
  • the containers have a capacity of at or about or at least at or about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, or 1000 mL capacity, such as between at or about 10 and at or about 100 or between at or about 10 and at or about 500 mL capacity, each inclusive.
  • the containers, e.g., bags are and/or are made from material which is stable and/or provide stable storage and/or maintenance of cells at one or more of various temperatures, such as in cold temperatures, e.g.
  • temperatures suitable for cryopreservation such as temperatures suitable for thawing the cells and body temperature such as at or about 37 °C, for example, to permit thawing, e.g., at the subject's location or location of treatment, e.g., at bedside, immediately prior to treatment.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container has one or more port, e.g., sterile access ports, for example, for connection of tubing or cannulation to one or more tubes, e.g., for intravenous or other infusion and/or for connection for purposes of transfer to and from other containers, such as cell culture and/or storage bags or other containers.
  • exemplary containers include infusion bags, intravenous solution bags, vials, including those with stoppers pierceable by a needle for injection.
  • the article of manufacture may further include a package insert or label with one or more pieces of identifying information and/or instructions for use.
  • the information or instructions indicates that the contents can or should be used to treat a particular condition or disease, and/or providing instructions therefor.
  • the label or package insert may indicate that the contents of the article of manufacture are to be used for treating the disease or condition.
  • the label or package insert provides instructions to treat a subject, e.g., the subject from which the cells have been derived, via a method involving the administration of a first and one or more consecutive doses of the cells, e.g., according to any of the instances of the disclosed methods.
  • the instructions specify administration, in a first dose, of one unit dose, e.g., the contents of a single individual container in the article of manufacture, followed by one or more consecutive doses at a specified time point or within a specified time window and/or after the detection of the presence or absence or amount or degree of one or more factors or outcomes in the subject.
  • the instructions specify administering one or more of the unit doses to the subject.
  • the label or package insert or packaging comprises an identifier to indicate the specific identity of the subject from which the cells are derived and/or are to be administered.
  • the identity of the subject from which the cells are derived is the same as the identity of the subject to which the cells are to be administered.
  • the identifying information may specify that the cells are to be administered to a particular patient, such as the one from which the cells were originally derived. Such information may be present in the packaging material and/or label in the form of a bar code or other coded identifier, or may indication the name and/or other identifying characteristics of the subject.
  • the article of manufacture in some instances includes one or more, typically a plurality, of containers containing compositions comprising the cells, e.g., individual unit dose forms thereof, and further include one or more additional containers with a composition contained therein which includes a further agent, such as a cytotoxic or otherwise therapeutic agent, for example, which is to be administered in combination, e.g., simultaneously or sequentially in any order, with the cells.
  • the article of manufacture may further include another or the same container comprising a pharmaceutically-acceptable buffer. It may further include other materials such as other buffers, diluents, filters, tubing, needles, and/or syringes.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
  • a "subject" is a mammal, such as a human or other animal, and typically is human.
  • the subject e.g., patient, to whom the immunomodulatory polypeptides, engineered cells, or compositions are administered, is a mammal, typically a primate, such as a human.
  • the primate is a monkey or an ape.
  • the subject can be male or female and can be any suitable age, including infant, juvenile, adolescent, adult, and geriatric subjects.
  • the subject is a non-primate mammal, such as a rodent.
  • treatment refers to complete or partial amelioration or reduction of a disease or condition or disorder, or a symptom, adverse effect or outcome, or phenotype associated therewith. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. The terms do not imply complete curing of a disease or complete elimination of any symptom or effect(s) on all symptoms or outcomes.
  • delay development of a disease means to defer, hinder, slow, retard, stabilize, suppress and/or postpone development of the disease (such as cancer). This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease. For example, a late stage cancer, such as development of metastasis, may be delayed.
  • Preventing includes providing prophylaxis with respect to the occurrence or recurrence of a disease in a subject that may be predisposed to the disease but has not yet been diagnosed with the disease.
  • the provided cells and compositions are used to delay development of a disease or to slow the progression of a disease.
  • a function or activity is to reduce the function or activity when compared to otherwise same conditions except for a condition or parameter of interest, or alternatively, as compared to another condition.
  • cells that suppress tumor growth reduce the rate of growth of the tumor compared to the rate of growth of the tumor in the absence of the cells.
  • an "effective amount" of an agent e.g., a pharmaceutical formulation, cells, or composition, in the context of administration, refers to an amount effective, at dosages/amounts and for periods of time necessary, to achieve a desired result, such as a therapeutic or prophylactic result.
  • a “therapeutically effective amount” of an agent refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result, such as for treatment of a disease, condition, or disorder, and/or pharmacokinetic or pharmacodynamic effect of the treatment.
  • the therapeutically effective amount may vary according to factors such as the disease state, age, sex, and weight of the subject, and the immunomodulatory polypeptides or engineered cells administered.
  • the disclosed methods involve administering the immunomodulatory polypeptides, engineered cells, or compositions at effective amounts, e.g., therapeutically effective amounts.
  • prophylactically effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • pharmaceutical formulation refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • nucleotides or amino acid positions "correspond to" nucleotides or amino acid positions in a disclosed sequence refers to nucleotides or amino acid positions identified upon alignment with the disclosed sequence to maximize identity using a standard alignment algorithm, such as the GAP algorithm.
  • aligning the sequences one can identify corresponding residues, for example, using conserved and identical amino acid residues as guides.
  • sequences of amino acids are aligned so that the highest order match is obtained (see, e.g.
  • amino acid substitution may include replacement of one amino acid in a polypeptide with another amino acid.
  • the substitution may be a conservative amino acid substitution or a non-conservative amino acid substitution.
  • Amino acid substitutions may be introduced into a binding molecule, e.g., antibody, of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased immunogenicity, or improved ADCC or CDC.
  • Amino acids generally can be grouped according to the following common side-chain properties:
  • conservative substitutions can involve the exchange of a member of one of these classes for another member of the same class.
  • non-conservative amino acid substitutions can involve exchanging a member of one of these classes for another class.
  • composition refers to any mixture of two or more products, substances, or compounds, including cells. It may be a solution, a suspension, liquid, powder, a paste, aqueous, non-aqueous or any combination thereof.
  • a statement that a cell or population of cells is "positive" for a particular marker refers to the detectable presence on or in the cell of a particular marker, typically a surface marker.
  • a surface marker refers to the presence of surface expression as detected by flow cytometry, for example, by staining with an antibody that specifically binds to the marker and detecting said antibody, wherein the staining is detectable by flow cytometry at a level substantially above the staining detected carrying out the same procedure with an isotype-matched control under otherwise identical conditions and/or at a level substantially similar to that for cell known to be positive for the marker, and/or at a level substantially higher than that for a cell known to be negative for the marker.
  • a statement that a cell or population of cells is "negative" for a particular marker refers to the absence of substantial detectable presence on or in the cell of a particular marker, typically a surface marker.
  • a surface marker refers to the absence of surface expression as detected by flow cytometry, for example, by staining with an antibody that specifically binds to the marker and detecting said antibody, wherein the staining is not detected by flow cytometry at a level substantially above the staining detected carrying out the same procedure with an isotype-matched control under otherwise identical conditions, and/or at a level substantially lower than that for cell known to be positive for the marker, and/or at a level substantially similar as compared to that for a cell known to be negative for the marker.
  • vector refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked.
  • the term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors.”
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
  • Example 1 Correlation Between Phenotype of Starting Composition and the Ratio of CD4+ to CD8+ CAR Expressing Cells in a CAR+ T cell Composition
  • CAR+ T cell compositions containing autologous T cells expressing a chimeric antigen receptor (CAR) were generated from apheresis collected from 11 separate donors with four test samples separately processed from each donor sample.
  • One donor was a patient with myeloma and the remaining donors were healthy subjects.
  • each sample was assessed by flow cytometry for cell viability using an apoptotic marker and for surface expression of CD4 and CD8 to determine the ratio of viable CD4+ to CD8+ T cells (CD4/CD8 ratio) in each apheresis sample run.
  • CD4+ and CD8+ T cells were selected from apheresis samples by immuno-affinity-based selection.
  • CD4+ T cells and CD8+ T cells were combined at a 1:1 ratio of viable CD4+ to CD8+ cells.
  • a sample of the combined cells was assessed by flow cytometry for cell viability using an apoptotic specific marker and for surface expression of markers that included CD4, CD8, CD45RA and CCR7.
  • the ratio of viable CD45RA+/CCR7+CD4+ to viable CD45RA+/CCR7+CD8+ (CD45RA+/CCR7+ CD4/CD8 ratio) in the mixture of selected CD4 and CD8 cells was determined.
  • the combined CD4+ and CD8+ T cells were activated by incubation with anti-CD3 and anti-CD28 antibody-coated beads in the presence of cytokines, and then were transduced with a lentiviral vector encoding an anti-BCMA CAR.
  • the CAR contained an scFv antigen-binding domain specific for BCMA, a spacer, a CD28 transmembrane region, a 4-1BB costimulatory signaling region, and a CD3-zeta derived intracellular signaling domain. After transduction, cells were expanded and then frozen by cryopreservation.
  • the cells in the frozen composition were thawed and assessed by flow cytometry for viability, surface expression of CD4 and CD8, and CAR expression using a BCMA-Fc reagent.
  • the ratio of viable CAR+ cells that were CD4+ to viable CAR+ cells that were CD8+ was determined (CAR+ CD4/CD8 ratio).
  • the degree of correlation of the mean ratios from each subject was assessed by bivariate analysis and the bivariate normal ellipse representing the 0.990 probability region for the plotted data are shown in in FIGS. 1A and 1B , respectively.
  • Table 1A and Table 1B display the results of a Pearson correlation analysis carried out with respect to the data plotted in FIG. 1A and FIG. 1B , respectively.
  • the CAR+ CD4/CD8 ratio in the final T cell composition positively correlated with the starting na ⁇ ve-like cell CD4/CD8 T cell ratio, as determined by the ratio of CD45RA+/CCR7+ CD4/CD8 ratio in the mixture of CD4 and CD8 cells.
  • the results in FIG. 1B show that the correlation between the ratio of CD45RA+/CCR7+/CD4+ cells to CD45RA+/CCR7+/CD8+ cells in the starting sample to the CAR+ CD4/CD8 ratio in the T cell composition is high based on Pearson correlation coefficient and p value ⁇ 0.0001. This correlation held despite variations in input composition and between process runs.
  • a CD45RA+/CCR7+/CD4+ to CD45RA+/CCR7+/CD8+ ratio of about 1.1:1 was determined to result in a CAR+ CD4/CD8 ratio of about 1: 1 in the output T cell composition.
  • Example 2 Correlation between phenotypes of the starting T cell population and the ratio of CAR+CD4+ to CAR+CD8+ T cells in engineered CAR+ T cell compositions
  • a total of 50 engineered CAR+ T cell compositions were generated from apheresis samples collected from 15 healthy donors and one multiple myeloma patient.
  • viable selected CD4+ and CD8+ T cells were combined into a starting cell composition at a 1:1 ratio and then activated, transduced, and expanded as described in Example 1.
  • Samples from the starting compositions of combined viable CD4+ and CD8+ T cells were assessed by flow cytometry for viability and for surface expression of markers that included CD4, CD8, CD27, CD45RA, CCR7, and CD62L.
  • Samples from the engineered CAR+ T cell compositions were assessed for CAR expression and for surface expression of markers that included CD4 and CD8. Averages were calculated for ratios of CD4+CAR+ T cells to CD8+CAR+T cells (CAR+ CD4/CD8 ratio) of individual CAR+ T cell compositions of the same donor that were generated using the same exemplary process as described in Example 1, except that certain process parameters were varied. The average CAR+ CD4/CD8 ratios were analyzed for correlations with various phenotypes of the starting compositions of combined viable CD4+ to CD8+ cells. Combining the selected CD4+ and CD8+ T-cells at 1:1 ratio prior to the activation did not necessarily correlate with a 1:1 CAR+ CD4/CD8 ratio in the generated output cell compositions.
  • the degree of the correlations between the average CAR+ CD4/CD8 ratios and the phenotypes from each donor were assessed by bivariate analysis.
  • the bivariate normal ellipses representing the 0.950 probability are shown for ratios for CD45RA+/CCR7+/CD4+ T cell to CD45RA+/CCR7+/CD8+ (CD45RA+/CCR7+ CD4/CD8 ratio; FIG. 2A ); CD62L-/CCR7+/CD4+ to CD62L-/CCR7+/CD8+ (CD62L-/CCR7+ CD4/CD8 ratio; FIG.
  • Table 2 displays the results of Pearson correlation analysis carried out with respect to the data plotted in FIGS. 2A-2C .
  • the CAR+ CD4/CD8 ratio was shown to correlate positively with CD62L-/CCR7+ CD4/CD8 and CD27+/CCR7+ CD4/CD8 ratios in both healthy donor and patient sample starting cell compositions. Based on the model fit, it was calculated that a starting CD27+/CCR7+ CD4/CD8 ratio of 1.69:1 would be predicted to generate an output cell composition with a CAR+ CD4/CD8 ratio of approximately 1:1.
  • Example 3 Process to Produce CAR+ T cell Composition Based on Phenotype of Starting Composition
  • CAR+ T cell compositions containing autologous T cells expressing a CAR were generated from apheresis collected from 3 separate donors, including two healthy donors and one multiple myeloma patient.
  • CD4+ and CD8+ T cells were selected from apheresis samples as described in Example 1. The apheresis samples and the selected CD4+ and CD8+ T cells were assessed by flow cytometry for viability and surface markers including CD27, CCR7, CD4, and CD8.
  • the frequency of CD27+/CCR7+ cells among the selected CD4+ and CD8+ T cells was determined, with each donor exhibiting a different ratio of CD27+/CCR7+ CD4+ cells to CD27+/CCR7+ CD8+ cells in apheresis samples.
  • the patient sample exhibited a ratio of CD27+/CCR7+ CD4+ cells to CD27+/CCR7+ CD8+ cells of approximately 12.2:1
  • the two healthy donor samples exhibited ratios of CD27+/CCR7+ CD4+ cells to CD27+/CCR7+ CD8+ cells of 3.56:1 and 2.15:1.
  • Input cell compositions were generated by either (1) combining selected CD4+ and CD8+ cells at a 1:1 viable CD4+ to CD8+ ratio (viable CD4/CD8) or (2) combining CD4+ and CD8+ cells at a 1.69:1 ratio of CD27+/CCR7+CD4+ cells and CD27+/CCR7+ CD8+ cells (CD27+/CCR7+ CD4/CD8) before activation .
  • a total of either 300 ⁇ 10 6 cells or 100 ⁇ 10 6 cells from the input compositions were activated, transduced, and expanded to generate output cell compositions substantially as described in Example 1.
  • the total number of cells in the activation step was not observed to impact the CAR+ CD4/CD8 ratio.
  • Output cell compositions generated from input compositions containing viable CD4/CD8 cells mixed at a 1:1 ratio showed grater variation in CAR+ CD4/CAR+ CD8 ratios, compared with compositions generated from input compositions containing CD4+ and CD8+ cells at a 1.69:1 ratio of CD27+/CCR7+ CD4+ cells and CD27+/CCR7+ CD8+ cells.
  • output compositions generated using patient material in an exemplary process exhibited CAR+ CD4/CAR+ CD8 ratios of approximately 7.6 and 8.6 when 100 ⁇ 10 6 cells or 300 ⁇ 10 6 cells were activated, respectively.
  • Example 4 Assessment of Samples From Diseased Subjects of Correlation Between Phenotype of Starting Composition and the Ratio of CD4+ to CD8+ CAR Expressing Cells in a CAR+ T Cell Composition
  • CAR+ T cell compositions containing autologous T cells expressing a chimeric antigen receptor (CAR) were generated from apheresis collected from 7 separate donors with multiple myeloma.
  • CD4+ and CD8+ T cells were selected from apheresis samples by immuno-affinity-based selection.
  • CD4+ T cells and CD8+ T cells were combined at a 1:1 ratio of viable CD4+ to CD8+ cells.
  • a sample of the combined cells was assessed for surface expression of markers that included CD4, CD8, CD27, CD45RA, CCR7 and CD62L.
  • CD27+/CCR7+ CD4+ cells to CD27+/CCR7+ CD8+ cells (CD27+/CCR7+ CD4/CD8 ratio)
  • CD45RA+/CCR7+ CD4+ cells to CD45RA+/CCR7+ CD8+ cells (CD45RA+/CCR7+ CD4/CD8 ratio)
  • CD62L-/CCR7+ CD4+ cells to CD62L-/CCR7+ CD8+ cells
  • a CAR+ T cell composition expressing an anti-BCMA CAR was generated using the process substantially as described in Example 1, including activation, transduction, expansion and cryopreservation of generated cells.
  • the cells in the frozen composition were thawed and assessed by flow cytometry for viability, surface expression of CD4 and CD8, and CAR expression using a BCMA-Fc reagent.
  • the ratio of viable CAR+ cells that were CD4+ to viable CAR+ cells that were CD8+ was determined (CAR+ CD4/CD8 ratio).
  • the degree of correlation of the mean ratios from each subject was assessed by bivariate normal ellipse representing the 0.950 probability region for the plotted data are shown in in FIGS. 3A-3C .
  • Tables 3A-3C display the results of correlation analysis carried out with respect to the data plotted in FIGS. 3A-3C . Significance ⁇ 0.05 is noted by *.
  • ESKYGPPCPPCP spacer (IgG4hinge) (aa) Homo sapiens 2 GAATCTAAGTACGGACCGCCCTGCCCCCCTTGCCCT spacer (IgG4hinge) (nt) Homo sapiens 3 Hinge-CH3 spacer Homo sapiens 4 Hinge-CH2-CH3 spacer Homo sapiens 5 IgD-hinge-Fc Homo sapiens 6 LEGGGEGRGSLLTCGDVEENPGPR T2A artificial 7 tEGFR artificial 8 FWVLVVVGGVLACYSLLVTVAFIIFWV CD28 (amino acids 153-179 of Accession No.
  • Homo sapiens 9 CD28 (amino acids 114-179 of Accession No. P10747) Homo sapiens 10 RSKRSRLLHSDYMNMTPRRPGPTRKHYQPYAPPRDFAAYRS CD28 (amino acids 180-220 of P10747) Homo sapiens 11 RSKRSRGGHSDYMNMTPRRPGPTRKHYQPYAPPRDFAAYRS CD28 (LL to GG) Homo sapiens 12 KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL 4-1BB (amino acids 214-255 of Q07011.1) Homo sapiens 13 CD3 zeta Homo sapiens 14 CD3 zeta Homo sapiens 15 CD3 zeta Homo sapiens 16 tEGFR artificial 17 EGRGSLLTCGDVEENPGP T2A artificial 18 PLGLWA MMP cleavable linker 19 GSGATNFSLLKQAGDVEENPGP P2A

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Hematology (AREA)
  • Mycology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Urology & Nephrology (AREA)
  • Developmental Biology & Embryology (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
EP18760159.6A 2017-08-09 2018-08-09 Methods for producing genetically engineered cell compositions and related compositions Active EP3664820B1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762543363P 2017-08-09 2017-08-09
US201762596770P 2017-12-08 2017-12-08
PCT/US2018/046149 WO2019032927A1 (en) 2017-08-09 2018-08-09 METHODS FOR PRODUCING GENETICALLY MODIFIED CELL COMPOSITIONS AND COMPOSITIONS THEREOF

Publications (2)

Publication Number Publication Date
EP3664820A1 EP3664820A1 (en) 2020-06-17
EP3664820B1 true EP3664820B1 (en) 2023-07-19

Family

ID=63405421

Family Applications (1)

Application Number Title Priority Date Filing Date
EP18760159.6A Active EP3664820B1 (en) 2017-08-09 2018-08-09 Methods for producing genetically engineered cell compositions and related compositions

Country Status (12)

Country Link
US (2) US11851678B2 (zh)
EP (1) EP3664820B1 (zh)
JP (2) JP7275104B2 (zh)
KR (1) KR20200054178A (zh)
CN (1) CN111246862A (zh)
AU (1) AU2018313950A1 (zh)
BR (1) BR112020001605A2 (zh)
CA (1) CA3070579A1 (zh)
ES (1) ES2959953T3 (zh)
MA (1) MA49979A (zh)
MX (1) MX2020001490A (zh)
WO (1) WO2019032927A1 (zh)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111225675B (zh) 2017-06-02 2024-05-03 朱诺治疗学股份有限公司 使用过继细胞疗法治疗的制品和方法
EP3720480A2 (en) * 2017-12-08 2020-10-14 Juno Therapeutics, Inc. Phenotypic markers for cell therapy and related methods
WO2021004400A1 (zh) * 2019-07-06 2021-01-14 苏州克睿基因生物科技有限公司 一种表达cd3抗体受体复合物的免疫细胞及其用途
JP2023504736A (ja) * 2019-12-06 2023-02-06 ジュノー セラピューティクス インコーポレイテッド Gprc5d標的結合ドメインに対する抗イディオタイプ抗体ならびに関連する組成物および方法
TW202140790A (zh) * 2020-01-22 2021-11-01 愛爾蘭商佧琺藥業有限公司 病毒載體轉導細胞的方法

Family Cites Families (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4452773A (en) 1982-04-05 1984-06-05 Canadian Patents And Development Limited Magnetic iron-dextran microspheres
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
US4690915A (en) 1985-08-08 1987-09-01 The United States Of America As Represented By The Department Of Health And Human Services Adoptive immunotherapy as a treatment modality in humans
US4795698A (en) 1985-10-04 1989-01-03 Immunicon Corporation Magnetic-polymer particles
US5219740A (en) 1987-02-13 1993-06-15 Fred Hutchinson Cancer Research Center Retroviral gene transfer into diploid fibroblasts for gene therapy
ES2067018T3 (es) 1988-12-28 1995-03-16 Stefan Miltenyi Procedimiento y materiales para la separacion en alto gradiente magnetico de materiales biologicos.
US5200084A (en) 1990-09-26 1993-04-06 Immunicon Corporation Apparatus and methods for magnetic separation
CA2096222C (en) 1990-11-13 1998-12-29 Stephen D. Lupton Bifunctional selectable fusion genes
EP0804590A1 (en) 1993-05-21 1997-11-05 Targeted Genetics Corporation Bifunctional selectable fusion genes based on the cytosine deaminase (cd) gene
US5827642A (en) 1994-08-31 1998-10-27 Fred Hutchinson Cancer Research Center Rapid expansion method ("REM") for in vitro propagation of T lymphocytes
WO1996013593A2 (en) 1994-10-26 1996-05-09 Procept, Inc. Soluble single chain t cell receptors
WO1996018105A1 (en) 1994-12-06 1996-06-13 The President And Fellows Of Harvard College Single chain t-cell receptor
US20020150914A1 (en) 1995-06-30 2002-10-17 Kobenhavns Universitet Recombinant antibodies from a phage display library, directed against a peptide-MHC complex
US6013516A (en) 1995-10-06 2000-01-11 The Salk Institute For Biological Studies Vector and method of use for nucleic acid delivery to non-dividing cells
US5773224A (en) 1996-02-12 1998-06-30 Grandics; Peter Immunoselection system for cell elution
DE19608753C1 (de) 1996-03-06 1997-06-26 Medigene Gmbh Transduktionssystem und seine Verwendung
US6451995B1 (en) 1996-03-20 2002-09-17 Sloan-Kettering Institute For Cancer Research Single chain FV polynucleotide or peptide constructs of anti-ganglioside GD2 antibodies, cells expressing same and related methods
JP2001519143A (ja) 1997-10-02 2001-10-23 スノル・モレキュラー・コーポレーション 可溶性の一本鎖t細胞レセプタータンパク質
US5994136A (en) 1997-12-12 1999-11-30 Cell Genesys, Inc. Method and means for producing high titer, safe, recombinant lentivirus vectors
RU2149023C1 (ru) 1998-04-20 2000-05-20 Ерхов Валентин Сергеевич Способ получения специфической антисыворотки к универсальному опухолевому антигену и способ диагностики злокачественных опухолей с использованием этой антисыворотки
JP2002515243A (ja) 1998-05-19 2002-05-28 アヴィデックス リミテッド 多価t細胞受容体複合体
EP1109921A4 (en) 1998-09-04 2002-08-28 Sloan Kettering Inst Cancer FOR PROSTATE-SPECIFIC MEMBRANE-ANTI-SPECIFIC FUSION RECEPTORS AND THEIR USE
US6410319B1 (en) 1998-10-20 2002-06-25 City Of Hope CD20-specific redirected T cells and their use in cellular immunotherapy of CD20+ malignancies
WO2000043551A1 (en) 1999-01-26 2000-07-27 Cornell Research Foundation, Inc. Determining viral load in double negative t cells
US20030235908A1 (en) 2000-02-24 2003-12-25 Xcyte Therapies, Inc. Activation and expansion of cells
US20040191260A1 (en) 2003-03-26 2004-09-30 Technion Research & Development Foundation Ltd. Compositions capable of specifically binding particular human antigen presenting molecule/pathogen-derived antigen complexes and uses thereof
CA2410510A1 (en) 2000-06-02 2001-12-13 Memorial Sloan-Kettering Cancer Center Artificial antigen presenting cells and methods of use thereof
US7446179B2 (en) 2000-11-07 2008-11-04 City Of Hope CD19-specific chimeric T cell receptor
US7070995B2 (en) 2001-04-11 2006-07-04 City Of Hope CE7-specific redirected immune cells
US20090257994A1 (en) 2001-04-30 2009-10-15 City Of Hope Chimeric immunoreceptor useful in treating human cancers
WO2009003492A1 (en) 2007-07-03 2009-01-08 Dako Denmark A/S Mhc multimers, methods for their generation, labeling and use
JP4317940B2 (ja) 2001-08-31 2009-08-19 イミュノコア・リミテッド 物質
US7939059B2 (en) 2001-12-10 2011-05-10 California Institute Of Technology Method for the generation of antigen-specific lymphocytes
US6992176B2 (en) 2002-02-13 2006-01-31 Technion Research & Development Foundation Ltd. Antibody having a T-cell receptor-like specificity, yet higher affinity, and the use of same in the detection and treatment of cancer, viral infection and autoimmune disease
EP1485075A4 (en) 2002-02-20 2006-04-26 Dyax Corp MHC-PEPTIDE COMPLEX BINDING LIGANDS
US20030170238A1 (en) 2002-03-07 2003-09-11 Gruenberg Micheal L. Re-activated T-cells for adoptive immunotherapy
US7446190B2 (en) 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
ES2375724T3 (es) 2002-09-27 2012-03-05 The General Hospital Corporation Dispositivo microflu�?dico para seperación de células y sus usos.
CA2501870C (en) 2002-10-09 2013-07-02 Avidex Limited Single chain recombinant t cell receptors
US20050129671A1 (en) 2003-03-11 2005-06-16 City Of Hope Mammalian antigen-presenting T cells and bi-specific T cells
WO2004096975A2 (en) 2003-05-02 2004-11-11 Insception Bioscience, Inc. Apparatus and methods for amplification of blood stem cell numbers
US20090304679A1 (en) 2004-05-27 2009-12-10 Weidanz Jon A Antibodies as T cell receptor mimics, methods of production and uses thereof
US20090226474A1 (en) 2004-05-27 2009-09-10 Weidanz Jon A Antibodies as T cell receptor mimics, methods of production and uses thereof
CA2567814C (en) 2004-05-27 2013-07-23 Jon A. Weidanz Antibodies as t cell receptor mimics, methods of production and uses thereof
DK1791865T3 (da) 2004-06-29 2010-11-01 Immunocore Ltd Celler der udtrykker en modificeret T-cellerecptor
WO2007117602A2 (en) 2006-04-07 2007-10-18 Biogen Idec Ma Inc. Isolation and use of human regulatory t cells
US20080085532A1 (en) 2006-09-18 2008-04-10 Jorn Gorlach Method for determining the immune status of a subject
ES2663323T3 (es) 2007-03-30 2018-04-12 Memorial Sloan-Kettering Cancer Center Expresión constitutiva de ligandos coestimuladores en linfocitos T transferidos de manera adoptiva
ES2374863T3 (es) 2007-12-07 2012-02-22 Miltenyi Biotec Gmbh Centrífuga para separar una muestra en por lo menos dos componentes.
US8479118B2 (en) 2007-12-10 2013-07-02 Microsoft Corporation Switching search providers within a browser search box
RS56844B1 (sr) 2007-12-11 2018-04-30 Univ North Carolina Chapel Hill Retrovirusni vektori sa modifikovanim polipurinskim nizom
US20120164718A1 (en) 2008-05-06 2012-06-28 Innovative Micro Technology Removable/disposable apparatus for MEMS particle sorting device
JP5173594B2 (ja) 2008-05-27 2013-04-03 キヤノン株式会社 管理装置、画像形成装置及びそれらの処理方法
US20110070581A1 (en) 2009-04-27 2011-03-24 Amit Gupta Separation of Leukocytes
CA2777053A1 (en) 2009-10-06 2011-04-14 The Board Of Trustees Of The University Of Illinois Human single-chain t cell receptors
EP4049674A1 (en) 2009-11-03 2022-08-31 City of Hope Truncated epidermal growth factor receptor (egfrt) for transduced t cell selection
MX347078B (es) 2010-12-09 2017-04-10 Univ Pennsylvania Uso de celulas t modificadas por receptor de antigeno quimerico para tratar cancer.
US9402865B2 (en) 2011-01-18 2016-08-02 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
WO2012129514A1 (en) 2011-03-23 2012-09-27 Fred Hutchinson Cancer Research Center Method and compositions for cellular immunotherapy
SG193956A1 (en) 2011-04-01 2013-11-29 Sloan Kettering Inst Cancer T cell receptor-like antibodies specific for a wt1 peptide presented by hla-a2
US8398282B2 (en) 2011-05-12 2013-03-19 Delphi Technologies, Inc. Vehicle front lighting assembly and systems having a variable tint electrowetting element
CN103827292B (zh) 2011-07-01 2018-01-26 安姆根有限公司 哺乳动物细胞培养
ES2682750T3 (es) 2011-07-18 2018-09-21 Iba Gmbh Método de tinción reversible de una célula diana
US9353161B2 (en) 2011-09-13 2016-05-31 Uti Limited Partnership Streptavidin mutein exhibiting reversible binding for biotin and streptavidin binding peptide tagged proteins
WO2013062365A2 (ko) 2011-10-26 2013-05-02 국립암센터 변이 ctla4 유전자 이입 t 세포 및 이를 포함하는 항암 면역치료용 조성물
MX359234B (es) 2011-11-11 2018-09-20 Hutchinson Fred Cancer Res Inmunoterapia de celulas t objetivadas en ciclina a1 para cancer.
EP2814846B1 (en) 2012-02-13 2020-01-08 Seattle Children's Hospital d/b/a Seattle Children's Research Institute Bispecific chimeric antigen receptors and therapeutic uses thereof
WO2013126726A1 (en) 2012-02-22 2013-08-29 The Trustees Of The University Of Pennsylvania Double transgenic t cells comprising a car and a tcr and their methods of use
SG10201606970UA (en) 2012-02-23 2016-10-28 Juno Therapeutics Gmbh Chromatographic isolation of cells and other complex biological materials
AU2013256159B2 (en) 2012-05-03 2018-08-09 Fred Hutchinson Cancer Center Enhanced affinity T cell receptors and methods for making the same
WO2014011996A1 (en) 2012-07-13 2014-01-16 The Trustees Of The University Of Pennsylvania Methods of assessing the suitability of transduced t cells for administration
SG10201701339RA (en) 2012-08-20 2017-03-30 Seattle Children S Hospital Dba Seattle Children S Res Inst Method and compositions for cellular immunotherapy
WO2014055668A1 (en) 2012-10-02 2014-04-10 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
JP5372297B1 (ja) 2012-12-20 2013-12-18 三菱電機株式会社 車載装置及びプログラム
CN105358576B (zh) 2013-02-20 2020-05-05 诺华股份有限公司 使用人源化抗EGFRvIII嵌合抗原受体治疗癌症
CN103305464B (zh) 2013-06-05 2015-04-15 南昌大学 直接分离cd4+和cd8+淋巴细胞的方法
US9108442B2 (en) 2013-08-20 2015-08-18 Ricoh Company, Ltd. Image forming apparatus
KR102487608B1 (ko) 2014-04-07 2023-01-12 노파르티스 아게 항-cd19 키메라 항원 수용체를 사용한 암의 치료
IL292038A (en) 2014-04-23 2022-06-01 Juno Therapeutics Inc Methods for enriching and producing immune cell populations for stress treatment
MX370018B (es) 2014-04-25 2019-11-28 Bluebird Bio Inc Metodos mejorados para la elaboracion de terapias celulares adoptivas.
DK3166968T3 (da) 2014-05-02 2021-12-06 Univ Pennsylvania Sammensætninger og fremgangsmåder til kimæriske autoantistofreceptor-T-celler
ES2781175T3 (es) 2014-07-31 2020-08-31 Novartis Ag Subconjunto optimizado de células T que contienen un receptor de antígeno quimérico
TWI805109B (zh) 2014-08-28 2023-06-11 美商奇諾治療有限公司 對cd19具專一性之抗體及嵌合抗原受體
CN113388036A (zh) 2014-12-05 2021-09-14 纪念斯隆-凯特琳癌症中心 靶向b-细胞成熟抗原的抗体及其用途
IL285101B2 (en) 2014-12-05 2024-03-01 Memorial Sloan Kettering Cancer Center G protein-coupled receptor-directed antibodies and methods of use
IL252621B1 (en) 2014-12-05 2024-02-01 Memorial Sloan Kettering Cancer Center G-protein receptor-directed chimeric antigen receptors and their use
EP3227432B1 (en) 2014-12-05 2023-10-11 Memorial Sloan Kettering Cancer Center Chimeric antigen receptors targeting b-cell maturation antigen and uses thereof
ES2876974T3 (es) 2015-04-07 2021-11-15 Novartis Ag Combinación de terapia con receptor de antígeno quimérico y derivados de amino pirimidina
CN114958764A (zh) 2015-04-08 2022-08-30 诺华股份有限公司 Cd20疗法、cd22疗法和与cd19嵌合抗原受体(car)表达细胞的联合疗法
MA45488A (fr) 2015-10-22 2018-08-29 Juno Therapeutics Gmbh Procédés, kits et appareil de culture de cellules
BR112018008648A2 (pt) 2015-10-30 2018-11-27 Univ California métodos de geração de células t a partir de células tronco e métodos imunoterapêuticos ao usar células t
BR112019017767A2 (pt) 2017-02-27 2020-04-07 Juno Therapeutics Inc composições, artigos de fabricação e métodos relacionados à dosagem em terapia celular
CA3070575A1 (en) 2017-08-09 2019-02-14 Juno Therapeutics, Inc. Methods and compositions for preparing genetically engineered cells
EP3720480A2 (en) 2017-12-08 2020-10-14 Juno Therapeutics, Inc. Phenotypic markers for cell therapy and related methods
WO2019113557A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Process for producing a composition of engineered t cells

Also Published As

Publication number Publication date
MX2020001490A (es) 2020-08-06
US20200181575A1 (en) 2020-06-11
EP3664820A1 (en) 2020-06-17
MA49979A (fr) 2020-06-17
RU2020109864A (ru) 2021-09-14
US11851678B2 (en) 2023-12-26
RU2020109864A3 (zh) 2022-01-10
CN111246862A (zh) 2020-06-05
JP7275104B2 (ja) 2023-05-17
KR20200054178A (ko) 2020-05-19
ES2959953T3 (es) 2024-02-29
WO2019032927A1 (en) 2019-02-14
JP2020532954A (ja) 2020-11-19
US20240076617A1 (en) 2024-03-07
JP2022160657A (ja) 2022-10-19
CA3070579A1 (en) 2019-02-14
AU2018313950A1 (en) 2020-02-13
BR112020001605A2 (pt) 2020-08-11

Similar Documents

Publication Publication Date Title
JP7299841B2 (ja) 細胞療法と免疫調節化合物の併用
US20190350978A1 (en) Production of engineered cells for adoptive cell therapy
US20200289565A1 (en) Combination of a cell therapy and a gamma secretase inhibitor
EP3664820B1 (en) Methods for producing genetically engineered cell compositions and related compositions
US20190298772A1 (en) Combination therapy of a t cell-based therapy and a btk inhibitor
KR20190104528A (ko) Car-t 세포들 투여를 결정하는 방법
US20200239910A1 (en) Methods and compositions for preparing genetically engineered cells
KR20200108277A (ko) 세포를 배양하기 위한 무혈청 배지 제형 및 이의 사용 방법
CA3108698A1 (en) Methods for assessing integrated nucleic acids
RU2790444C2 (ru) Способы получения композиций генно-инженерных клеток и родственных композиций
US20240168012A1 (en) Methods of determining potency of a therapeutic cell composition
RU2795454C2 (ru) Способы и композиции для получения генно-инженерных клеток
RU2780156C2 (ru) Продуцирование сконструированных клеток для адоптивной клеточной терапии
KR20220146480A (ko) T 세포 형질도입 방법

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20200306

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20201124

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40032621

Country of ref document: HK

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20230131

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230525

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602018053641

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20230816

Year of fee payment: 6

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20230913

Year of fee payment: 6

Ref country code: IE

Payment date: 20230809

Year of fee payment: 6

Ref country code: GB

Payment date: 20230810

Year of fee payment: 6

Ref country code: ES

Payment date: 20230905

Year of fee payment: 6

Ref country code: CH

Payment date: 20230902

Year of fee payment: 6

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG9D

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20230821

Year of fee payment: 6

Ref country code: DE

Payment date: 20230816

Year of fee payment: 6

Ref country code: BE

Payment date: 20230818

Year of fee payment: 6

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1588800

Country of ref document: AT

Kind code of ref document: T

Effective date: 20230719

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231020

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231119

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231120

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231019

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231119

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20231020

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2959953

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20240229

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20230809

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20230809

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

VS25 Lapsed in a validation state [announced via postgrant information from nat. office to epo]

Ref country code: MD

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230719

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT